Next Article in Journal
Quantitative Characterization of Olaparib in Nanodelivery System and Target Cell Compartments by LC-MS/MS
Next Article in Special Issue
Traceless Solid-Phase Synthesis of Ketones via Acid-Labile Enol Ethers: Application in the Synthesis of Natural Products and Derivatives
Previous Article in Journal
Vasodilation Elicited by Isoxsuprine, Identified by High-Throughput Virtual Screening of Compound Libraries, Involves Activation of the NO/cGMP and H2S/KATP Pathways and Blockade of α1-Adrenoceptors and Calcium Channels
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Green and Facile Assembly of Diverse Fused N-Heterocycles Using Gold-Catalyzed Cascade Reactions in Water

1
Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu 610052, China
2
School of Biological Science and Technology, University of Jinan, Jinan 250022, China
3
State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Shanghai 201203, China
4
University of Chinese Academy of Sciences, Beijing 100049, China
*
Authors to whom correspondence should be addressed.
Submission received: 4 February 2019 / Revised: 1 March 2019 / Accepted: 4 March 2019 / Published: 11 March 2019
(This article belongs to the Special Issue Modern Strategies for Heterocycle Synthesis)

Abstract

:
The present study describes an AuPPh3Cl/AgSbF6-catalyzed cascade reaction between amine nucleophiles and alkynoic acids in water. This process proceeds in high step economy with water as the sole coproduct, and leads to the generation of two rings, together with the formation of three new bonds in a single operation. This green cascade process exhibits valuable features such as low catalyst loading, good to excellent yields, high efficiency in bond formation, excellent selectivity, great tolerance of functional groups, and extraordinarily broad substrate scope. In addition, this is the first example of the generation of an indole/thiophene/pyrrole/pyridine/naphthalene/benzene-fused N-heterocycle library through gold catalysis in water from readily available materials. Notably, the discovery of antibacterial molecules from this library demonstrates its high quality and potential for the identification of active pharmaceutical ingredients.

Graphical Abstract

1. Introduction

Rapid advances in genomics and proteomics have resulted in the identification of an increasing number of novel therapeutic targets [1,2,3,4,5], and the existing compound libraries can no longer well meet the needs of drug screening. Therefore, it is highly demanding to develop robust synthetic methods to construct new compound libraries for drug discovery aimed at these targets [6,7,8]. Considering the structurally diverse targets in a wide “biological space”, high-throughput screening (HTS) of skeletally diverse compounds, which occupy a broader “chemical space”, can apparently enhance the hit rates [9,10]. In addition to skeletal diversity, drug-like properties of the compounds are equally important for the generation of high-quality compound libraries [11,12,13,14], which can increase the possibility of identifying drug-like hit compounds. As a result, privileged structures have received wide attention in drug discovery because they are widely found in natural and pharmaceutical products [15,16,17,18,19]. Although privileged substructure-based diversity-oriented synthesis (pDOS) provides a useful access to assemble compound libraries with high-quality [20,21,22,23,24,25,26], it is still challenging to develop efficient and practical approaches to generate a variety of molecular frameworks embedded with privileged structures, especially in a green and sustainable manner. With our interests to develop green and efficient protocols to synthesize valuable N-heterocycles [27,28,29,30,31], we herein construct a library of privileged substructure-based N-heterocycles with diverse scaffolds using gold-catalyzed cascade reactions in water. To the best of our knowledge, this is the first example of the generation of pDOS compound library encompassing skeletal diversity, molecular complexity, and drug-like properties through gold catalysis in water.
Alkynoic acids are extensively used to react with amine nucleophiles to furnish heterocyclic compounds because of the efficient cascade reaction developed by Dixon’s group [32], in which, an activated cyclic enol lactone species, which derives from alkynoic acids, is involved as the key intermediate (Scheme 1a). Dixon and co-workers disclosed linear aliphatic terminal alkynoic acids reacted smoothly with amine nucleophiles bearing a nucleophilic carbon atom in toluene or xylene to produce pyrrole- or indole-based heterocyclic frameworks catalyzed by AuPPh3Cl/AgOTf. It should be noted that, based on Dixon’s pioneering work, the reactions of alkynoic acids with amine nucleophiles in aprotic solvents such as toluene, xylene, 1,2-dichloroethane, dichloromethane, etc., have been well studied by Patil’s group as well as our group [33,34,35,36,37,38,39,40,41,42,43]. However, the reactivity of alkynoic acids and amine nucleophiles in the environmentally friendly, abundant, and cheap solvent—water—was seldom explored [44,45], mainly because the ring opening reaction of the activated enol lactone intermediate in water at elevated temperature may lead to the failure of the cascade reaction. This reason prevented researchers’ steps from investigating the nature of the cascade reaction in water. To date, only two successful examples in water were reported by our group [44,45], although the alkynoic acids were only limited to linear aliphatic terminal alkynoic acids, amine nucleophiles were only limited to amine nucleophiles carrying a nucleophilic carbon atom, and complicated Au catalysts were required (Scheme 1b). However, considering water often displays unique reactivity and selectivity which can’t be obtained in common organic solvents [46,47,48,49,50,51] and it is an environmentally benign solvent, we aim to develop a more general cascade process between various alkynoic acids and amine nucleophiles in water with great interest. Despite the possibility of hydrolysis of enol lactone intermediate in water at high temperature which may result in the failure of the cascade reaction, we hypothesize that a more general cascade process could also be achieved in water but with a proper catalytic system. In the present study, we develop a greener, and more general and efficient catalytic system (AuPPh3Cl/AgSbF6/CF3CO2H) in water, which tolerates a broader substrate scope of alkynoic acids as well as amine nucleophiles. As shown in Scheme 1c, not only linear aliphatic terminal and internal alkynoic acids but also cyclic aromatic terminal and internal alkynoic acids are well tolerated. In addition, extraordinarily broad amine nucleophiles bearing a nucleophilic carbon/nitrogen/oxygen atom turned out to be suitable substrates. Besides, the reaction mechanism in water was carefully checked and studied for the first time. Interestingly, when D2O was used as the reaction solvent, we observed the highly deuterated products generated from the H–D exchange between the reaction substrates/intermediates and D2O. Herein, we also present the library construction of skeletally diverse N-heterocycles embedded with privileged structures employing different alkynoic acids and various amine nucleophiles as the building blocks. To our delight, five antimicrobial compounds were identified from this library after biological evaluation. The production of N-heterocycles with diverse scaffolds and the discovery of active pharmaceutical ingredients (APIs) demonstrate the power of this method in both organic synthesis and medicinal chemistry.

2. Results and Discussion

4-pentynoic acid (1a) and tryptamine (2a) were employed as the model substrates to optimize the cascade reaction conditions (Table 1). Pleasingly, treatment of starting materials 1a and 2a in water without any catalyst at 100 °C for 24 h gave the desired product SF1a, albeit with a low yield (entry 1). Then various metal catalysts were screened to improve the reaction yield. A screening of Pd, Cu, Ni, and Mn complexes (entries 2–11), disclosed, at first, that Pd(II) sources, such as PdCl2(CH3CN)2 and Pd(PPh3)2Cl2, NiOAc and Mn(OAc)2, had little enhancement on the yield; while Pd(0) sources such as Pd(PPh3)4 and Pd2(dba)3; copper catalysts, such as Cu(OAc)2 and Cu(OH)2, and CuCl; and Ni(PPh3)2Cl inhibited this transformation. A subsequent survey of Ag, Ru, and Co catalysts (entries 12–16) revealed that Ag2CO3, AgOAc, [RuCl2(p-cym)]2, CoCl2, and Co(acac)2 could obviously increase the yield, affording the product SF1a in moderate yields (38–57%). Notably, a good yield (70%) was achieved with AuPPh3Cl (entry 17), and increasing the temperature to 120 °C was much more efficient in improving the yield as compared with extending the reaction time to 48 h (entries 18 and 19). Encouraged by this result, an investigation of other Au catalysts at 120 °C was carried out (entries 20–24). Among them, none displayed higher catalytic reactivity than AuPPh3Cl. To further improve the yield of the product, a series of silver salts, which were proved to be able to increase the catalytic reactivity of gold catalysts [52,53,54,55,56,57], were screened as the additives (entries 25–28). To our delight, AgSbF6 was found to be the best choice, with which product SF1a was obtained in 91% yield (entry 28). In addition, other typical organic solvents were also tested as the reaction solvents, and toluene, xylene, and DCE also turned out to be suitable solvents, in which similar high isolated yields were observed (See Supplementary Materials for details). However, considering water is more environmentally benign, we then decided to use water as the solvent to explore the substrate scope of this method. In this way, the optimal reaction conditions were identified using a catalytic system consisting of AuPPh3Cl/AgSbF6 in water at 120 °C for 24 h.
After determining the optimal reaction conditions, we then began to construct a high-quality library of privileged substructure-based N-heterocycles with diverse scaffolds. We first examined the generality of the process with various amine nucleophiles 2 containing a nucleophilic carbon on a heteroaromatic or aromatic ring. In general, this process tolerated a variety of amine nucleophiles 2 and alkynoic acids 1, and 26 scaffolds embedded with privileged structures were furnished in good to high yields in water under optimal or modified reaction conditions (Scheme 2). For example, tryptamines reacted smoothly with terminal alkynoic acids such as 4-pentynoic acid under standard conditions to give products SF1a and SF1b in high yields. The reactions of tryptamines with other terminal alkynoic acids, such as 5-hexynoic acid, 2-ethynylbenzoic acid, and 2-(2-ethynylphenyl)acetic acid, also afforded the corresponding products (SF2a, SF2b, SF3a, SF3b, and SF4a) in good to high yields under a modified two-step one-pot process, in which CF3CO2H (TFA) was added to promote the iminium ion formation. In addition, internal alkynoic acids, such as 5-phenylpent-4-ynoic acid, 6-phenylhex-5-ynoic acid, 2-(phenylethynyl)benzoic acid, and 2-(2-(phenylethynyl)phenyl)acetic acid, were also tested as the substrates. Unfortunately, they all failed to react with tryptamines to give the desired products (SF1c, SF2c, SF3c, and SF4b) even under further improved conditions. Interestingly, 2-(1H-indol-2-yl)ethylamines could undergo this transformation with terminal alkynoic acids as well as internal alkynoic acids to yield the desired products SF5SF8 in 35–96% yields. It should be noted that no N1 ring closure products were observed when 2-(1H-indol-2-yl)ethylamines were used as the amine nucleophiles, suggesting excellent selectivity of this cascade process. This may be because the C3 nucleophilicity is stronger than N1 nucleophilicity [58]. Likewise, the reactivity of 2-(1H-indol-1-yl)ethanamines in this cascade reaction was very similar to that of tryptamines. They reacted well with terminal alkynoic acids while their reactions with internal alkynoic acids failed to give the desired products (SF9SF12). Interestingly, the protocol was also compatible with 3-(1H-indol-1-yl)propan-1-amines, which furnished products SF13 carrying a seven-membered ring in 54–80% yields. Similarly, products SF14SF16 were obtained in 59–86% yields when 2-(1H-indol-1-yl)anilines and alkynoic acids were subjected to the modified conditions. Notably, the indole-containing polycyclic frameworks, represented by compounds SF1SF16, are regarded as valuable N-heterocycles considering their ubiquitous presence in biologically active molecules [59,60,61,62,63]. Subsequently, 2-(1H-pyrrol-2-yl)ethanamines, 2-(1H-pyrrol-1-yl)ethanamines, 2-(thiophen-2-yl)ethanamines, and 2-(thiophen-3-yl)ethanamines were employed as amine nucleophiles. Their reactions with diverse alkynoic acids took place successfully to provide pyrrole- or thiophene-fused compounds SF17SF23 in moderate to high yields, despite the fact that stronger conditions were required. In particular, 2-phenylethanamines with electron-donating substituents on the benzene ring were also well tolerated, leading to the formation of benzene-fused heterocyclic products SF24SF26 in yields ranging from 47 to 88%. It is also worth noting that excellent selectivity was achieved in the reactions of 2-(1H-pyrrol-2-yl)ethanamine, 2-(thiophen-3-yl)ethanamine or 2-phenylethanamines, even though two potential cyclization sites existed in the final step. It should be noted that compound SF26 is the analog of tetrahydroberberines, which were extracted from the Chinese herb Corydalis ambigua and exhibited a broad range of biological activities [64,65].
To further broaden the substrate scope of this approach, amine nucleophiles 3 containing a nucleophilic heteroatom (Z = amide/aniline N, acid/alcohol O) were tested as substrates. Overall, this protocol was also applicable to diverse amine nucleophiles 3, and 17 scaffolds were constructed with high efficiency (Scheme 3). For instance, the reactions of 2-aminobenzamides with various alkynoic acids worked successfully, affording benzene-fused polycyclic products SF27SF30 in moderate to high yields. Gratifyingly, this approach was compatible with 2-(aminomethyl)anilines and benzene-1,2-diamines, although the desired benzene-based heterocyclic products SF31SF34 were obtained in lower yields. Remarkably, in the case of substrates such as 2-aminobenzamides and 2-(aminomethyl)anilines, which contain two nitrogen atoms as the nucleophiles, the nitrogen atom with stronger nucleophilicity tended to attack the enol lactone intermediate and therefore the other nitrogen atom with weaker nucleophilicity attacked the iminium ion intermediate to selectively provide the corresponding products, while not in the reverse way. Besides, 2-aminobenzoic acids, 3-amino-2-naphthoic acids, or 2-aminonicotinic acids reacted with various alkynoic acids smoothly, producing the corresponding benzene-, naphthalene-, or pyridine-fused heterocyclic products SF35SF41 in yields of 50–96%. Surprisingly, 2-aminobenzyl alcohols were also found to be suitable substrates, which could undergo the cascade reaction with alkynoic acids to give the desired benzene-fused polycyclic products SF42 and SF43, albeit with lower yields. In addition, we also tried to synthesize the indole-fused compounds SF44SF46 embedded with an eight- or nine-membered ring; unfortunately, we failed. This may be attributed to the low reactivities of the amine nucleophiles and the instability of the large rings in energetics.
The diversity of the library can be further expanded by the derivatization of the target compounds. We herein introduce the derivatization of the target compounds based on the simple reduction of the carbonyl group, and the selected results are shown in Scheme 4. Nine different scaffolds represented by indole-, pyrrole-, or thiophene-based polycyclic compounds SF47SF55 containing a tertiary amine were produced conveniently through an easy reduction of the corresponding precursors with LiAlH4/AlCl3. Notably, these scaffolds are very similar to those found in natural and pharmaceutical agents [65]. We expect the screening of these compounds towards specific biological targets might lead to the identification of bioactive molecules.
Thus, using various amine nucleophiles and alkynoic acids as the building blocks, a library of privileged substructure-based N-heterocycles with diverse scaffolds was constructed through gold catalysis in a green and efficient manner. It is worth noting this cascade process constructs three new bonds together with two rings in one chemical process, suggesting the high efficiency of this cascade reaction in synthesizing nitrogen-containing heterocyclic compounds. Regarding the large occurrence of nitrogen-containing heterocyclic compounds in APIs [66,67,68], the method presented in this paper is prospective since it could provide an environmentally benign and useful platform for the preparation of diverse nitrogen-containing heterocyclic compounds.
To verify our expectation that this approach could provide useful scaffolds with attractive bioactivities, a bioactivity study of this library was carried out. An initial pharmacological study of these nitrogen-containing heterocyclic compounds led to the discovery of five antimicrobial compounds—SF9d, SF29b, SF33, SF36, and SF41. The minimal inhibitory concentration (MIC) results revealed that compound SF36 displayed the most potent antibacterial activity against S. aureus strain, with a MIC90 value of 10–25 μg/mL (Table 2) (Time-kill assays and colony-forming unit studies of compounds SF9d, SF29b, SF33, SF36 and SF41 could be found in Supplementary Materials).
Mechanistic studies were carried out with deuterium-labeling experiments, and the hydrogens of the products were assigned at first by the analysis of the 1H-NMR, 13C-NMR, HSQC, HMBC, and 1H-1H COSY spectrum to confirm the deuterated positions (See Supplementary Materials for details). Interestingly, the reaction of 4-pentynoic acid 1a with tryptamine 2a in D2O under the standard conditions afforded the deuterated product [D]n-SF1a, not only at the methyl position but also at the β-position of the carbonyl (Scheme 5a). Specifically, a 96% deuteration at the methyl position and a similar deuteration (90%) of the two unequal hydrogens at the β-position of the carbonyl were observed. The reactions of 2-(1H-indol-2-yl)ethylamines with 4-pentynoic acid 1a in D2O gave the similar results (Scheme 5b,c).
According to the results of deuterium-labeling experiments, we hypothesize two possible reaction pathways (Scheme 6 and Scheme 7). The reaction of with 4-pentynoic acid 1a with tryptamine 2a in the presence of Au catalyst in D2O is taken as the example to illustrate the reaction pathway. The first hypothetic reaction pathway (Scheme 6) may involve the gold-catalyzed hydration of carbon–carbon triple bond, which was observed in our previous work [69]. The H–D exchange between the carboxyl group of 4-pentynoic acid 1a and D2O leads to the formation of intermediate A1. Gold-catalyzed addition of D2O to the carbon–carbon triple bond of intermediate A1 produces intermediate A2, which undergoes two keto–enol tautomerizations to give intermediate A4. The subsequent H–D exchange between the hydroxyl group of intermediate A4 and D2O affords intermediate A5, which undergoes keto–enol tautomerization again to give intermediate A6. The two acidic protons at the α-position of the carbonyl in intermediate A6 undergo H–D exchange with D2O via keto–enol tautomerizations to yield intermediate A7. The following condensation between intermediate A7 and tryptamine 2a, the subsequent iminium ion formation, and the final cyclization achieve the product [D]n-SF1a. The second hypothetic reaction pathway is shown in Scheme 7; the H–D exchange between the carboxyl group of 4-pentynoic acid 1a and D2O produces intermediate A1. Gold-catalyzed intramolecular cyclization of intermediate A1 produces enol lactone intermediate B1, which is attacked by tryptamine 2a to give intermediate B2. The subsequent H–D exchange between the hydroxyl group of intermediate B2 and D2O yields intermediate B3, which undergoes enol–keto tautomerization to provide intermediate B4. Similarly, the three acidic protons at the α-position of the carbonyl in intermediate B4 undergo H–D exchange with D2O via keto–enol tautomerizations to yield intermediate C1, which is converted into the product [D]n-SF1a via an iminium ion formation/cyclization sequence.
To further verify the reaction mechanism, the reaction of 4-pentynoic acid 1a and tryptamine 2a under gold catalysis in O18-labeled water was carried out first. This reaction in H2O18 was stopped after 0.5 h to track the reaction intermediates. As shown in Scheme 8a, apart from the remaining starting materials, intermediate 2a″ and the product SF1a were obtained in 21% and 15% yield, respectively. While the O18-labeled intermediate 2a′ was not observed. This result clearly indicates the hydration of alkyne moiety, which is proposed in Scheme 6, is not involved. By contrast, the commercially purchased enol lactone D reacted smoothly with tryptamine 2a without the gold catalyst (Scheme 8b). This result shows that the enol lactone species B1, which is proposed in Scheme 7, is likely to be the key intermediate.
On the basis of the above results of mechanistic experiments, a final proposed reaction mechanism is outlined in Scheme 9. The proposed mechanism commences with the coordination of the gold catalyst to the carbon–carbon triple bond of alkynoic acids to produce intermediate I1. The subsequent intramolecular exo cyclization of I1 yields intermediate I2. The following protodemetalation of intermediate I2 takes place to produce the enol lactone species I3 with the regeneration of the catalyst. Then intermediate I3 undergoes aminolysis by amine nucleophiles to give intermediate I4, which tautomerizes to produce intermediate I5. Intermediate I5 is converted into the iminium ion I8 under the catalysis of the gold catalyst. The final nucleophilic cyclization of intermediate I8 affords the desired products with the release of the gold catalyst. The stronger nucleophilicity of amine nucleophiles compared to that of H2O results in the aminolysis of enol lactone I3 by amine nucleophiles instead of hydrolysis by H2O. It should be noted that the reaction solvent H2O participates in this cascade reaction via the H–H exchange with the carboxyl group of alkynoic acids, the hydroxyl group of intermediate I4, and the α hydrogen atoms of the carbonyl group of intermediate I5, as demonstrated by deuterium-labeling experiments. Besides, TFA could promote this reaction by accelerating the formation of iminium ion I8.

3. Materials and Methods

3.1. General Information

If not otherwise specified, the starting materials were obtained from commercial sources and used directly without purification. Analytical thin-layer chromatography (TLC): HSGF 254 (0.15–0.2 mm thickness). Detection under UV light at 254 nm. Column chromatography: Separations were carried out on silica gel FCP 200–300. Yields refer to isolated compounds. Melting point apparatus: a micro melting point apparatus, values are uncorrected. Nuclear magnetic resonance (NMR) apparatus: a Brucker instrument. Chemical shifts (δ) are given in ppm. Proton coupling patterns were recorded as singlet (s), doublet (d), triplet (t), quartet (q), and multiplet (m). LRMS (low-resolution mass) and HRMS (high-resolution mass) were measured on a spectrometer with an electrospray ionization (ESI) source.

3.2. General Procedure for the Preparation of Compounds SF1a, SF1b, SF5a, SF5b, and SF5c

A suspension of alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 (0.5 mmol), and AuPPh3Cl/AgSbF6 (0.005 mmol) in H2O (4.0 mL) was stirred at 120 °C for 24 h. At ambient temperature, saturated Na2CO3 solution (25.0 mL) was added to the reaction mixture. The resulting mixture was then extracted with ethyl acetate (3 × 15 mL). The combined organic layers were washed with brine, and dried over Na2SO4. After filtration and removal of the solvents in vacuo, the crude product was purified by flash chromatography on silica gel to provide the desired product.
11b-Methyl-5,6,11,11b-tetrahydro-1H-indolizino[8,7-b]indol-3(2H)-one (SF1a): white solid (109.7 mg, yield 91%), mp 260–261 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.54 (s, 3H), 2.07–1.99 (m, 1H), 2.32–2.20 (m, 2H), 2.66–2.55 (m, 2H), 2.75–2.67 (m, 1H), 3.11–3.00 (m, 1H), 4.25–4.16 (m, 1H), 7.01–6.94 (m, 1H), 7.10-7.02 (m, 1H), 7.32 (d, J = 8.0 Hz, 1H), 7.39 (d, J = 7.8 Hz, 1H), 11.06 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.9 (CO), 139.0 (C, Ar), 135.9 (C, Ar), 126.3 (C, Ar), 121.0 (CH, Ar), 118.6 (CH, Ar), 118.0 (CH, Ar), 111.1 (CH, Ar), 104.7 (C, Ar), 58.9 (C), 34.3 (CH2), 32.6 (CH2), 30.1 (CH2), 25.0 (CH3), 20.9 (CH2); ESI-LRMS m/z: 241 [M + H]+; ESI-HRMS m/z calcd for M + H+ 241.1335, found: 241.1331. The characterization data is in accordance with that reported in [32].
8-Methoxy-11b-methyl-5,6,11,11b-tetrahydro-1H-indolizino[8,7-b]indol-3(2H)-one (SF1b): yellow oil (120.2 mg, yield 89%). 1H-NMR (500 MHz, DMSO-d6) δ 1.53 (s, 3H), 2.07–1.97 (m, 1H), 2.31–2.20 (m, 2H), 2.64–2.52 (m, 2H), 2.73–2.64 (m, 1H), 3.10–2.98 (m, 1H), 3.74 (s, 3H), 4.24–4.15 (m, 1H), 6.70 (dd, J = 8.7, 2.4 Hz, 1H), 6.89 (d, J = 2.3 Hz, 1H), 7.20 (d, J = 8.7 Hz, 1H), 10.88 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 171.9 (CO), 153.2 (C, Ar), 139.7 (C, Ar), 130.9 (C, Ar), 126.6 (C, Ar), 111.7 (CH, Ar), 110.8 (CH, Ar), 104.6 (C, Ar), 100.2 (CH, Ar), 58.9 (C), 55.4 (OCH3), 34.3 (CH2), 32.6 (CH2), 30.1 (CH2), 25.0 (CH3), 21.0 (CH2); ESI-LRMS m/z: 271 [M + H]+; ESI-HRMS m/z calcd for M + H+ 271.1441, found: 271.1437. The characterization data is in accordance with that reported in [43].
11c-Methyl-5,6,7,11c-tetrahydro-1H-indolizino[7,8-b]indol-3(2H)-one (SF5a): yellow solid (114.2 mg, yield 95%), mp 96–97 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.54 (s, 3H), 2.02–1.92 (m, 1H), 2.27–2.17 (m, 1H), 2.54–2.47 (m, 1H), 2.65–2.54 (m, 1H), 2.81–2.67 (m, 2H), 3.16–3.04 (m, 1H), 4.27–4.18 (m, 1H), 7.00–6.92 (m, 1H), 7.08–7.00 (m, 1H), 7.30 (d, J = 8.0 Hz, 1H), 7.47 (d, J = 7.8 Hz, 1H), 10.90 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.5 (CO), 135.9 (C, Ar), 130.5 (C, Ar), 123.9 (C, Ar), 120.5 (CH, Ar), 118.6 (CH, Ar), 117.9 (CH, Ar), 115.3 (C, Ar), 111.1 (CH, Ar), 59.2 (C), 33.2 (CH2), 33.2 (CH2), 30.2 (CH2), 25.0 (CH3), 22.8 (CH2); ESI-LRMS m/z: 241 [M + H]+; ESI-HRMS m/z calcd for M + H+ 241.1335, found: 241.1332. The characterization data is in accordance with that reported in [43].
10-Methoxy-11c-methyl-5,6,7,11c-tetrahydro-1H-indolizino[7,8-b]indol-3(2H)-one (SF5b): pale yellow solid (129.5 mg, yield 96%), mp 193–194 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.54 (s, 3H), 2.01–1.91 (m, 1H), 2.27–2.18 (m, 1H), 2.64–2.52 (m, 2H), 2.79–2.65 (m, 2H), 3.14–3.03 (m, 1H), 3.77 (s, 3H), 4.25–4.15 (m, 1H), 6.69 (dd, J = 8.7, 2.4 Hz, 1H), 6.93 (d, J = 2.3 Hz, 1H), 7.19 (d, J = 8.7 Hz, 1H), 10.72 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.5 (CO), 153.1 (C, Ar), 131.3 (C, Ar), 131.0 (C, Ar), 124.2 (C, Ar), 115.1 (C, Ar), 111.7 (CH, Ar), 109.8 (CH, Ar), 100.6 (CH, Ar), 59.2 (C), 55.5 (OCH3), 33.2 (CH2), 33.0 (CH2), 30.1 (CH2), 24.8 (CH3), 22.9 (CH2); ESI-LRMS m/z: 271 [M + H]+; ESI-HRMS m/z calcd for M + H+ 271.1441, found: 271.1437. The characterization data is in accordance with that reported in [43].
10,11c-Dimethyl-5,6,7,11c-tetrahydro-1H-indolizino[7,8-b]indol-3(2H)-one (SF5c): pale yellow oil (118.4 mg, yield 93%). 1H-NMR (500 MHz, DMSO-d6) δ 1.53 (s, 3H), 2.02–1.91 (m, 1H), 2.29–2.17 (m, 1H), 2.38 (s, 3H), 2.50–2.46 (m, 1H), 2.64–2.54 (m, 1H), 2.80–2.65 (m, 2H), 3.15–3.03 (m, 1H), 4.26–4.15 (m, 1H), 6.86 (dd, J = 8.2, 1.0 Hz, 1H), 7.18 (d, J = 8.2 Hz, 1H), 7.25 (s, 1H), 10.75 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 171.5 (CO), 134.2 (C, Ar), 130.5 (C, Ar), 127.1 (C, Ar), 124.1 (C, Ar), 122.0 (CH, Ar), 117.6 (CH, Ar), 114.8 (C, Ar), 110.8 (CH, Ar), 59.2 (C), 33.3 (CH2), 33.2 (CH2), 30.2 (CH2), 25.0 (CH3), 22.8 (CH2), 21.3 (CH3); ESI-LRMS m/z: 255 [M + H]+; ESI-HRMS m/z calcd for M + H+ 255.1492, found: 255.1489. The characterization data is in accordance with that reported in [43].

3.3. General Procedure for the Preparation of Compounds SF1c, SF2SF4, SF5d, SF5e and SF6SF46

A suspension of alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 or 3 (0.5 mmol), and AuPPh3Cl/AgSbF6 (with the amount indicated) in H2O (4.0 mL) was stirred at the temperature indicated for 20 h. Then the reaction mixture was cooled to room temperature, and CF3COOH (0.5 mmol) was added, and the resulting mixture was stirred for another 4 h at the temperature indicated. At ambient temperature, saturated Na2CO3 solution (25.0 mL) was added to the reaction mixture. The resulting mixture was then extracted with ethyl acetate (3 × 15 mL). The combined organic layers were washed with brine, and dried over Na2SO4. After filtration and removal of the solvents in vacuo, the crude product was purified by flash chromatography on silica gel to yield the desired product.
12b-Methyl-1,2,3,6,7,12b-hexahydroindolo[2,3-a]quinolizin-4(12H)-one (SF2a): white solid (82.6 mg, yield 65%), mp 255–256 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.60 (s, 3H), 1.80–1.68 (m, 2H), 1.97–1.86 (m, 1H), 2.32–2.21 (m, 1H), 2.45–2.32 (m, 2H), 2.61–2.53 (m, 1H), 2.69–2.62 (m, 1H), 2.99–2.87 (m, 1H), 4.90–4.81 (m, 1H), 7.00–6.93 (m, 1H), 7.10–7.02 (m, 1H), 7.32 (d, J = 8.0 Hz, 1H), 7.39 (d, J = 7.8 Hz, 1H), 10.92 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 167.9 (CO), 139.7 (C, Ar), 136.0 (C, Ar), 126.2 (C, Ar), 120.9 (CH, Ar), 118.5 (CH, Ar), 117.9 (CH, Ar), 111.1 (CH, Ar), 105.8 (C, Ar), 56.4 (C), 35.6 (CH2), 34.8 (CH2), 31.8 (CH2), 25.3 (CH3), 21.0 (CH2), 16.3 (CH2); ESI-LRMS m/z: 255 [M + H]+; ESI-HRMS m/z calcd for M + H+ 255.1492, found: 255.1488. The characterization data is in accordance with that reported in [43].
9-Methoxy-12b-methyl-1,2,3,6,7,12b-hexahydroindolo[2,3-a]quinolizin-4(12H)-one (SF2b): pale yellow solid (95.4 mg, yield 67%), mp 190–191 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.59 (s, 3H), 1.79–1.66 (m, 2H), 1.97–1.84 (m, 1H), 2.42–2.21 (m, 3H), 2.66–2.51 (m, 2H), 2.99–2.86 (m, 1H), 3.74 (s, 3H), 4.90–4.79 (m, 1H), 6.70 (dd, J = 8.7, 2.4 Hz, 1H), 6.89 (d, J = 2.4 Hz, 1H), 7.20 (d, J = 8.7 Hz, 1H), 10.73 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 167.9 (CO), 153.2 (C, Ar), 140.4 (C, Ar), 131.0 (C, Ar), 126.5 (C, Ar), 111.7 (CH, Ar), 110.7 (CH, Ar), 105.7 (C, Ar), 100.1 (CH, Ar), 56.4 (C), 55.4 (OCH3), 35.6 (CH2), 34.8 (CH2), 31.8 (CH2), 25.4 (CH3), 21.1 (CH2), 16.3 (CH2); ESI-LRMS m/z: 285 [M + H]+; ESI-HRMS m/z calcd for M + H+ 285.1598, found: 285.1593. The characterization data is in accordance with that reported in [43].
13b-Methyl-7,8,13,13b-tetrahydro-5H-benzo[1,2]indolizino[8,7-b]indol-5-one (SF3a): white solid (119.4 mg, yield 83%), mp 283–284 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.86 (s, 3H), 2.75–2.63 (m, 1H), 2.85–2.75 (m, 1H), 3.47–3.36 (m, 1H), 4.59–4.47 (m, 1H), 7.03–6.93 (m, 1H), 7.15–7.05 (m, 1H), 7.44–7.34 (m, 2H), 7.58–7.49 (m, 1H), 7.79–7.68 (m, 2H), 8.32 (d, J = 7.9 Hz, 1H), 11.35 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 167.2 (CO), 149.3 (C, Ar), 136.2 (C, Ar), 135.2 (C, Ar), 132.2 (CH, Ar), 130.3 (C, Ar), 128.6 (CH, Ar), 126.0 (C, Ar), 123.2 (CH, Ar), 122.8 (CH, Ar), 121.6 (CH, Ar), 118.9 (CH, Ar), 118.3 (CH, Ar), 111.2 (CH, Ar), 106.3 (C, Ar), 62.0 (C), 35.4 (CH2), 25.9 (CH3), 21.4 (CH2); ESI-LRMS m/z: 289 [M + H]+; ESI-HRMS m/z calcd for M + H+ 289.1335, found: 289.1330. The characterization data is in accordance with that reported in [43].
10-Methoxy-13b-methyl-7,8,13,13b-tetrahydro-5H-benzo[1,2]indolizino[8,7-b]indol-5-one (SF3b): white solid (146.9 mg, yield 92%), mp 164–165 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.84 (s, 3H), 2.72–2.60 (m, 1H), 2.81–2.73 (m, 1H), 3.45–3.35 (m, 1H), 3.73 (s, 3H), 4.57–4.45 (m, 1H), 6.73 (dd, J = 8.7, 2.1 Hz, 1H), 6.89 (d, J = 2.1 Hz, 1H), 7.26 (d, J = 8.7 Hz, 1H), 7.58–7.47 (m, 1H), 7.77–7.67 (m, 2H), 8.29 (d, J = 7.9 Hz, 1H), 11.18 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 167.2 (CO), 153.3 (C, Ar), 149.4 (C, Ar), 135.8 (C, Ar), 132.2 (CH, Ar), 131.2 (C, Ar), 130.2 (C, Ar), 128.6 (CH, Ar), 126.3 (C, Ar), 123.2 (CH, Ar), 122.8 (CH, Ar), 111.9 (CH, Ar), 111.5 (CH, Ar), 106.2 (C, Ar), 100.3 (CH, Ar), 62.0 (C), 55.4 (OCH3), 35.5 (CH2), 26.0 (CH3), 21.5 (CH2); ESI-LRMS m/z: 319 [M + H]+; ESI-HRMS m/z calcd for M + H+ 319.1441, found: 319.1435. The characterization data is in accordance with that reported in [43].
14b-Methyl-8,9,14,14b-tetrahydroindolo[2′,3′:3,4]pyrido[2,1-a]isoquinolin-6(5H)-one (SF4a): pale yellow solid (120.7 mg, yield 80%), mp 137–138 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.84 (s, 3H), 2.50–2.41 (m, 1H), 2.86–2.75 (m, 1H), 2.96–2.86 (m, 1H), 3.63 (d, J = 19.4 Hz, 1H), 4.08 (d, J = 19.2 Hz, 1H), 4.96–4.85 (m, 1H), 7.08–6.98 (m, 1H), 7.18–7.12 (m, 1H), 7.24–7.18 (m, 1H), 7.31–7.24 (m, 2H), 7.52–7.43 (m, 3H), 11.56 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ169.0 (CO), 139.9 (C, Ar), 136.1 (C, Ar), 135.1 (C, Ar), 132.3 (C, Ar), 127.8 (CH, Ar), 127.5 (CH, Ar), 126.4 (CH, Ar), 126.0 (C, Ar), 124.1 (CH, Ar), 121.4 (CH, Ar), 118.8 (CH, Ar), 118.2 (CH, Ar), 111.3 (CH, Ar), 109.2 (C, Ar), 60.9 (C), 38.0 (CH2), 37.9 (CH2), 26.2 (CH3), 21.0 (CH2); ESI-LRMS m/z: 303 [M + H]+; ESI-HRMS m/z calcd for M + H+ 303.1492, found: 303.1487. The characterization data is in accordance with that reported in [43].
10-Fluoro-11c-methyl-5,6,7,11c-tetrahydro-1H-indolizino[7,8-b]indol-3(2H)-one (SF5d): pale yellow solid (95.1 mg, yield 74%), mp 104–105 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.52 (s, 3H), 1.99–1.87 (m, 1H), 2.28–2.16 (m, 1H), 2.64–2.51 (m, 2H), 2.83–2.67 (m, 2H), 3.16–3.01 (m, 1H), 4.28–4.15 (m, 1H), 6.93–6.82 (m, 1H), 7.34–7.21 (m, 2H), 11.00 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.5 (CO), 156.7 (d, JC–F = 231.1 Hz, CF, Ar), 132.9 (C, Ar), 132.5 (C, Ar), 124.0 (d, JC–F = 10.1 Hz, C, Ar), 115.7 (d, JC–F = 4.5 Hz, C, Ar), 111.9 (d, JC–F = 9.9 Hz, CH, Ar), 108.3 (d, JC–F = 25.8 Hz, CH, Ar), 102.9 (d, JC–F = 23.4 Hz, CH, Ar), 59.0 (C), 33.1 (CH2), 32.9 (CH2), 30.1 (CH2), 24.8 (CH3), 22.9 (CH2); ESI-LRMS m/z: 259 [M + H]+; ESI-HRMS m/z calcd for M + H+ 259.1241, found: 259.1238. The characterization data is in accordance with that reported in [43].
11c-Benzyl-5,6,7,11c-tetrahydro-1H-indolizino[7,8-b]indol-3(2H)-one (SF5e): pale yellow oil (69.4 mg, yield 44%). 1H-NMR (500 MHz, CDCl3) δ 1.78–1.65 (m, 1H), 2.22–2.09 (m, 2H), 2.72–2.60 (m, 2H), 2.97–2.85 (m, 1H), 3.08–2.98 (m, 1H), 3.21 (d, J = 13.9 Hz, 1H), 3.30 (d, J = 13.9 Hz, 1H), 4.50 (dd, J = 12.9, 6.4 Hz, 1H), 7.12–7.06 (m, 2H), 7.22–7.13 (m, 2H), 7.26–7.23 (m, 3H), 7.35 (d, J = 8.0 Hz, 1H), 7.52 (d, J = 7.6 Hz, 1H), 8.33 (s, 1H); 13C-NMR (125 MHz, CDCl3) δ 174.3 (CO), 136.7 (C, Ar), 136.2 (C, Ar), 130.6 (C, Ar), 130.2 (2 × CH, Ar), 128.5 (2 × CH, Ar), 127.0 (CH, Ar), 124.4 (C, Ar), 121.8 (CH, Ar), 119.9 (CH, Ar), 118.5 (CH, Ar), 115.9 (C, Ar), 111.3 (CH, Ar), 63.8 (C), 44.6 (CH2), 34.3 (CH2), 31.6 (CH2), 31.0 (CH2), 23.1 (CH2); ESI-LRMS m/z: 317 [M + H]+; ESI-HRMS m/z calcd for M + H+ 317.1648, found: 317.1651. The characterization data is in accordance with that reported in [43].
12c-Methyl-1,2,3,6,7,12c-hexahydroindolo[3,2-a]quinolizin-4(8H)-one (SF6a): white solid (86.8 mg, yield 68%), mp 132–133 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.63 (s, 3H), 1.77–1.65 (m, 2H), 2.01–1.85 (m, 1H), 2.32–2.19 (m, 1H), 2.43–2.32 (m, 1H), 2.78–2.60 (m, 3H), 3.01–2.89 (m, 1H), 4.92–4.80 (m, 1H), 6.99–6.90 (m, 1H), 7.07–6.99 (m, 1H), 7.29 (d, J = 8.0 Hz, 1H), 7.51 (d, J = 7.8 Hz, 1H), 10.91 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 168.1 (CO), 136.1 (C, Ar), 131.9 (C, Ar), 124.0 (C, Ar), 120.3 (CH, Ar), 118.6 (CH, Ar), 118.5 (CH, Ar), 115.7 (C, Ar), 111.1 (CH, Ar), 57.0 (C), 35.6 (CH2), 34.7 (CH2), 31.9 (CH2), 25.1 (CH3), 23.3 (CH2), 16.5 (CH2); ESI-LRMS m/z: 255 [M + H]+; ESI-HRMS m/z calcd for M + H+ 255.1492, found: 255.1488. The characterization data is in accordance with that reported in [43].
12c-Benzyl-1,2,3,6,7,12c-hexahydroindolo[3,2-a]quinolizin-4(8H)-one (SF6b): pale yellow oil (57.9 mg, yield 35%). 1H-NMR (400 MHz, DMSO-d6) δ 1.52–1.41 (m, 1H), 1.68–1.54 (m, 1H), 1.90–1.78 (m, 1H), 2.24–2.05 (m, 2H), 2.61–2.53 (m, 1H), 2.81–2.64 (m, 3H), 3.24 (d, J = 13.6 Hz, 1H), 3.41 (d, J = 13.5 Hz, 1H), 4.85–4.74 (m, 1H), 6.94–6.86 (m, 1H), 7.06–6.96 (m, 3H), 7.23–7.14 (m, 3H), 7.29 (d, J = 8.0 Hz, 1H), 7.33 (d, J = 7.9 Hz, 1H), 10.95 (s, 1H); 13C-NMR (150 MHz, DMSO-d6) δ 169.2 (CO), 137.8 (C, Ar), 136.0 (C, Ar), 133.0 (C, Ar), 130.4 (2 × CH, Ar), 127.9 (2 × CH, Ar), 126.4 (CH, Ar), 124.3 (C, Ar), 120.2 (CH, Ar), 119.0 (CH, Ar), 118.5 (CH, Ar), 114.2 (C, Ar), 111.1 (CH, Ar), 60.9 (C), 44.3 (CH2), 35.0 (CH2), 33.9 (CH2), 31.7 (CH2), 23.0 (CH2), 16.3 (CH2); ESI-LRMS m/z: 331 [M + H]+; ESI-HRMS m/z calcd for M + H+ 331.1805, found: 331.1812. The characterization data is in accordance with that reported in [43].
13b-Methyl-6,7-dihydro-5H-benzo[1,2]indolizino[7,8-b]indol-9(13bH)-one (SF7a): pale yellow solid (116.4 mg, yield 81%), mp 235–236 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.90 (s, 3H), 2.80–2.70 (m, 1H), 2.91–2.80 (m, 1H), 3.45–3.37 (m, 1H), 4.56–4.46 (m, 1H), 7.12–7.03 (m, 2H), 7.34–7.26 (m, 1H), 7.52–7.44 (m, 1H), 7.69–7.64 (m, 1H), 7.71 (d, J = 7.5 Hz, 1H), 8.12–8.03 (m, 1H), 8.27 (d, J = 7.8 Hz, 1H), 11.10 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 167.3 (CO), 151.4 (C, Ar), 135.9 (C, Ar), 132.3 (C, Ar), 132.0 (CH, Ar), 130.2 (C, Ar), 128.1 (CH, Ar), 124.3 (C, Ar), 123.2 (CH, Ar), 123.1 (CH, Ar), 120.7 (CH, Ar), 119.0 (CH, Ar), 119.0 (CH, Ar), 111.6 (C, Ar), 111.3 (CH, Ar), 63.6 (C), 34.6 (CH2), 25.7 (CH3), 23.4 (CH2); ESI-LRMS m/z: 289 [M + H]+; ESI-HRMS m/z calcd for M + H+ 289.1335, found: 289.1330. The characterization data is in accordance with that reported in [43].
13b-Benzyl-6,7-dihydro-5H-benzo[1,2]indolizino[7,8-b]indol-9(13bH)-one (SF7b): white solid (121.1 mg, yield 66%), mp 281–282 °C. 1H-NMR (500 MHz, DMSO-d6) δ 2.91–2.72 (m, 2H), 3.49–3.38 (m, 1H), 3.57 (d, J = 13.8 Hz, 1H), 3.85 (d, J = 13.9 Hz, 1H), 4.53 (dd, J = 13.0, 5.6 Hz, 1H), 6.95–6.87 (m, 2H), 7.08–6.98 (m, 3H), 7.20–7.08 (m, 2H), 7.39–7.30 (m, 2H), 7.46 (d, J = 7.4 Hz, 1H), 7.66–7.58 (m, 1H), 8.32 (d, J = 7.8 Hz, 1H), 8.43 (d, J = 7.8 Hz, 1H), 11.14 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 167.7 (CO), 148.8 (C, Ar), 135.9 (C, Ar), 135.7 (C, Ar), 132.8 (C, Ar), 131.4 (CH, Ar), 131.0 (C, Ar), 130.0 (2 × CH, Ar), 127.8 (CH, Ar), 127.3 (2 × CH, Ar), 126.2 (CH, Ar), 124.4 (C, Ar), 123.9 (CH, Ar), 122.6 (CH, Ar), 120.7 (CH, Ar), 119.4 (CH, Ar), 119.1 (CH, Ar), 111.3 (CH, Ar), 111.1 (C, Ar), 67.1 (C), 42.7 (CH2), 34.8 (CH2), 23.4 (CH2); ESI-LRMS m/z: 365 [M + H]+; ESI-HRMS m/z calcd for M + H+ 365.1648, found: 365.1649. The characterization data is in accordance with that reported in [43].
13b-Pentyl-6,7-dihydro-5H-benzo[1,2]indolizino[7,8-b]indol-9(13bH)-one (SF7c): pale yellow oil (68.7 mg, yield 40%). 1H-NMR (400 MHz, CDCl3) δ 0.77 (t, J = 6.7 Hz, 3H), 0.89–0.83 (m, 1H), 1.20–1.02 (m, 5H), 2.24–2.10 (m, 1H), 2.76–2.58 (m, 2H), 3.13–2.98 (m, 1H), 3.38–3.24 (m, 1H), 4.77 (dd, J = 13.1, 6.1 Hz, 1H), 7.23–7.12 (m, 2H), 7.31 (d, J = 7.8 Hz, 1H), 7.45–7.38 (m, 1H), 7.62–7.54 (m, 1H), 7.85 (d, J = 7.5 Hz, 1H), 7.99 (d, J = 7.7 Hz, 1H), 8.06 (d, J = 7.7 Hz, 1H), 8.23 (s, 1H); 13C-NMR (150 MHz, CDCl3) δ 169.4 (CO), 149.6 (C, Ar), 136.1 (C, Ar), 132.0 (CH, Ar), 131.9 (C, Ar), 131.8 (C, Ar), 128.1 (CH, Ar), 124.9 (C, Ar), 123.9 (CH, Ar), 122.9 (CH, Ar), 121.7 (CH, Ar), 120.0 (CH, Ar), 119.4 (CH, Ar), 113.5 (C, Ar), 111.4 (CH, Ar), 67.4 (C), 38.2 (CH2), 35.3 (CH2), 31.8 (CH2), 24.1 (CH2), 23.1 (CH2), 22.5 (CH2), 14.1 (CH3); ESI-LRMS m/z: 345 [M + H]+; ESI-HRMS m/z calcd for M + H+ 345.1961, found: 345.1966. The characterization data is in accordance with that reported in [43].
14c-Methyl-8,9,10,14c-tetrahydroindolo[3′,2’:3,4]pyrido[2,1-a]isoquinolin-6(5H)-one (SF8a): yellow solid (129.8 mg, yield 86%), mp 250–251 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.89 (s, 3H), 2.72–2.60 (m, 1H), 2.84–2.73 (m, 1H), 3.02–2.89 (m, 1H), 3.62 (d, J = 19.4 Hz, 1H), 4.10 (d, J = 19.2 Hz, 1H), 4.95–4.86 (m, 1H), 7.15–7.04 (m, 3H), 7.30–7.19 (m, 2H), 7.44–7.38 (m, 2H), 7.59 (d, J = 7.6 Hz, 1H), 11.26 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 169.3 (CO), 141.4 (C, Ar), 136.0 (C, Ar), 134.5 (C, Ar), 132.4 (C, Ar), 127.7 (CH, Ar), 127.1 (CH, Ar), 126.2 (C, Ar), 126.1 (CH, Ar), 124.7 (CH, Ar), 120.5 (CH, Ar), 120.2 (CH, Ar), 119.2 (CH, Ar), 111.5 (CH, Ar), 111.2 (C, Ar), 61.5 (C), 38.2 (CH2), 36.9 (CH2), 25.7 (CH3), 23.3 (CH2); ESI-LRMS m/z: 303 [M + H]+; ESI-HRMS m/z calcd for M + H+ 303.1492, found: 303.1486. The characterization data is in accordance with that reported in [43].
14c-Benzyl-8,9,10,14c-tetrahydroindolo[3’,2’:3,4]pyrido[2,1-a]isoquinolin-6(5H)-one (SF8b): pale yellow oil (66.5 mg, yield 35%). 1H-NMR (500 MHz, CDCl3) δ 1.84–1.73 (m, 1H), 2.48–2.38 (m, 1H), 2.67–2.56 (m, 1H), 3.40 (d, J = 13.8 Hz, 1H), 3.96–3.76 (m, 3H), 4.79 (dd, J = 12.7, 4.3 Hz, 1H), 6.80–6.71 (m, 2H), 7.08–7.00 (m, 2H), 7.16–7.08 (m, 2H), 7.28–7.20 (m, 2H), 7.33–7.28 (m, 2H), 7.50–7.42 (m, 1H), 7.66 (d, J = 7.9 Hz, 1H), 7.96–7.88 (m, 1H), 8.36 (s, 1H); 13C-NMR (125 MHz, CDCl3) δ 170.6 (CO), 141.0 (C, Ar), 136.4 (C, Ar), 136.1 (C, Ar), 135.7 (C, Ar), 132.3 (C, Ar), 130.4 (2 × CH, Ar), 128.2 (2 × CH, Ar), 128.0 (CH, Ar), 127.7 (CH, Ar), 127.3 (C, Ar), 126.9 (CH, Ar), 126.8 (CH, Ar), 126.0 (CH, Ar), 121.6 (CH, Ar), 121.4 (CH, Ar), 120.5 (CH, Ar), 111.7 (CH, Ar), 109.9 (C, Ar), 66.5 (C), 45.0 (CH2), 39.1 (CH2), 38.7 (CH2), 23.3 (CH2); ESI-LRMS m/z: 379 [M + H]+; ESI-HRMS m/z calcd for M + H+ 379.1805, found: 379.1815. The characterization data is in accordance with that reported in [43].
12b-Methyl-1,5,6,12b-tetrahydropyrrolo[2’,1’:3,4]pyrazino[1,2-a]indol-3(2H)-one (SF9a): white solid (107.8 mg, yield 90%), mp 121–122 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.59 (s, 3H), 2.33–2.17 (m, 2H), 2.47–2.36 (m, 1H), 2.65–2.53 (m, 1H), 3.49–3.38 (m, 1H), 3.81–3.68 (m, 1H), 4.34–4.22 (m, 2H), 6.34 (s, 1H), 7.08–7.01 (m, 1H), 7.15–7.09 (m, 1H), 7.38 (d, J = 8.1 Hz, 1H), 7.51 (d, J = 7.7 Hz, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 172.1 (CO), 142.1 (C, Ar), 135.2 (C, Ar), 127.6 (C, Ar), 120.7 (CH, Ar), 119.9 (CH, Ar), 119.8 (CH, Ar), 109.6 (CH, Ar), 95.5 (CH, Ar), 58.6 (C), 40.9 (CH2), 34.3 (CH2), 33.6 (CH2), 29.7 (CH2), 27.2 (CH3); ESI-LRMS m/z: 241 [M + H]+; ESI-HRMS m/z calcd for M + H+ 241.1335, found: 241.1330. The characterization data is in accordance with that reported in [43].
11,12b-Dimethyl-1,5,6,12b-tetrahydropyrrolo[2’,1’:3,4]pyrazino[1,2-a]indol-3(2H)-one (SF9b): pale yellow oil (111.3 mg, yield 88%). 1H-NMR (300 MHz, CDCl3) δ 1.65 (s, 3H), 2.49–2.37 (m, 3H), 2.54 (s, 3H), 2.73–2.57 (m, 1H), 3.47–3.32 (m, 1H), 3.96–3.81 (m, 1H), 4.20 (dd, J = 11.7, 4.7 Hz, 1H), 4.52 (dd, J = 13.6, 5.1 Hz, 1H), 6.30 (s, 1H), 6.98–6.89 (m, 1H), 7.15–7.09 (m, 2H); 13C-NMR (100 MHz, CDCl3) δ 173.2 (CO), 141.1 (C, Ar), 135.4 (C, Ar), 130.0 (C, Ar), 127.9 (C, Ar), 121.8 (CH, Ar), 120.7 (CH, Ar), 106.8 (CH, Ar), 94.8 (CH, Ar), 59.5 (C), 41.3 (CH2), 35.0 (CH2), 34.3 (CH2), 30.4 (CH2), 27.8 (CH3), 18.8 (CH3); ESI-LRMS m/z: 255 [M + H]+; ESI-HRMS m/z calcd for M + H+ 255.1492, found: 255.1488. The characterization data is in accordance with that reported in [45].
12b-Methyl-3-oxo-1,2,3,5,6,12b-hexahydropyrrolo[2’,1’:3,4]pyrazino[1,2-a]indole-11-carbonitrile (SF9c): yellow oil (104.3 mg, yield 79%). 1H-NMR (300 MHz, CDCl3) δ 1.66 (s, 3H), 2.54–2.32 (m, 3H), 2.73–2.59 (m, 1H), 3.49–3.33 (m, 1H), 4.02–3.88 (m, 1H), 4.26 (dd, J = 11.7, 4.8 Hz, 1H), 4.56 (dd, J = 13.7, 5.2 Hz, 1H), 6.52 (s, 1H), 7.26–7.19 (m, 1H), 7.52–7.44 (m, 2H); 13C-NMR (125 MHz, CDCl3) δ 173.1 (CO), 144.6 (C, Ar), 135.4 (C, Ar), 129.7 (C, Ar), 125.7 (CH, Ar), 121.2 (CH, Ar), 118.7 (CN), 113.9 (CH, Ar), 102.8 (C, Ar), 95.6 (CH, Ar), 59.3 (C), 41.5 (CH2), 34.8 (CH2), 33.9 (CH2), 30.2 (CH2), 27.6 (CH3); ESI-LRMS m/z: 266 [M + H]+; ESI-HRMS m/z calcd for M + H+ 266.1288, found: 266.1282. The characterization data is in accordance with that reported in [45].
9-Fluoro-12b-methyl-1,5,6,12b-tetrahydropyrrolo[2’,1’:3,4]pyrazino[1,2-a]indol-3(2H)-one (SF9d): pale yellow solid (43.6 mg, yield 68%), mp 71–72 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.58 (s, 3H), 2.30–2.18 (m, 2H), 2.44–2.36 (m, 1H), 2.63–2.52 (m, 1H), 3.49–3.38 (m, 1H), 3.77–3.67 (m, 1H), 4.32–4.22 (m, 2H), 6.36 (s, 1H), 6.94–6.86 (m, 1H), 7.26 (dd, J = 10.2, 2.2 Hz, 1H), 7.50 (dd, J = 8.6, 5.4 Hz, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 172.1 (CO), 158.5 (d, JC–F = 234.3 Hz, CF, Ar), 142.9 (d, JC–F = 3.7 Hz, C, Ar), 135.2 (d, JC–F = 12.5 Hz, C, Ar), 124.2 (C, Ar), 120.9 (d, JC–F = 10.0 Hz, CH, Ar), 108.1 (d, JC–F = 24.3 Hz, CH, Ar), 96.3 (d, JC–F = 26.3 Hz, CH, Ar), 95.6 (CH, Ar), 58.6 (C), 41.1 (CH2), 34.3 (CH2), 33.5 (CH2), 29.6 (CH2), 27.3 (CH3); ESI-LRMS m/z: 259 [M + H]+; ESI-HRMS m/z calcd for M + H+ 259.1241, found: 259.1238.
10-Chloro-12b-methyl-1,5,6,12b-tetrahydropyrrolo[2’,1’:3,4]pyrazino[1,2-a]indol-3(2H)-one (SF9e): pale yellow oil (87.5 mg, yield 64%). 1H-NMR (300 MHz, CDCl3) δ 1.63 (s, 3H), 2.52–2.32 (m, 3H), 2.71–2.56 (m, 1H), 3.45–3.32 (m, 1H), 3.94–3.81 (m, 1H), 4.17 (dd, J = 11.6, 4.6 Hz, 1H), 4.52 (dd, J = 13.7, 5.0 Hz, 1H), 6.23 (s, 1H), 7.20–7.11 (m, 2H), 7.52 (s, 1H); 13C-NMR (100 MHz, CDCl3) δ 173.1 (CO), 143.0 (C, Ar), 134.1 (C, Ar), 129.1 (C, Ar), 126.1 (C, Ar), 121.8 (CH, Ar), 119.9 (CH, Ar), 110.1 (CH, Ar), 96.0 (CH, Ar), 59.4 (C), 41.3 (CH2), 34.8 (CH2), 34.0 (CH2), 30.2 (CH2), 27.7 (CH3); ESI-LRMS m/z: 277 ([M + H]+, Cl37), 275 ([M + H]+, Cl35); ESI-HRMS m/z calcd for M + H+ 275.0946, found: 275.0943. The characterization data is in accordance with that reported in [45].
13b-Methyl-2,3,6,7-tetrahydro-1H-pyrido[2’,1’:3,4]pyrazino[1,2-a]indol-4(13bH)-one (SF10a): white solid (73.5 mg, yield 58%), mp 113–114 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.67 (s, 3H), 1.78–1.69 (m, 1H), 1.98–1.88 (m, 1H), 2.07–1.99 (m, 1H), 2.42–2.26 (m, 3H), 3.40–3.25 (m, 1H), 3.81–3.70 (m, 1H), 4.33–4.24 (m, 1H), 4.95–4.85 (m, 1H), 6.34 (s, 1H), 7.06–7.01 (m, 1H), 7.14–7.07 (m, 1H), 7.37 (d, J = 8.1 Hz, 1H), 7.50 (d, J = 7.8 Hz, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 168.1 (CO), 142.4 (C, Ar), 135.1 (C, Ar), 127.7 (C, Ar), 120.6 (CH, Ar), 119.8 (CH, Ar), 119.7 (CH, Ar), 109.4 (CH, Ar), 95.2 (CH, Ar), 56.7 (C), 41.2 (CH2), 36.6 (CH2), 34.5 (CH2), 31.7 (CH2), 28.2 (CH3), 16.9 (CH2); ESI-LRMS m/z: 255 [M + H]+; ESI-HRMS m/z calcd for M + H+ 255.1492, found: 255.1488. The characterization data is in accordance with that reported in [43].
12,13b-Dimethyl-2,3,6,7-tetrahydro-1H-pyrido[2’,1’:3,4]pyrazino[1,2-a]indol-4(13bH)-one (SF10b): colorless oil (95.4 mg, yield 71%). 1H-NMR (300 MHz, CDCl3) δ 1.73 (s, 3H), 2.06–1.86 (m, 2H), 2.26–2.11 (m, 1H), 2.50–2.32 (m, 2H), 2.65–2.51 (m, 4H), 3.39–3.23 (m, 1H), 4.01–3.87 (m, 1H), 4.22–4.10 (m, 1H), 5.16 (dd, J = 13.7, 4.5 Hz, 1H), 6.26 (s, 1H), 6.99–6.88 (m, 1H), 7.18–7.07 (m, 2H); 13C-NMR (100 MHz, CDCl3) δ 169.5 (CO), 141.4 (C, Ar), 135.2 (C, Ar), 129.9 (C, Ar), 127.9 (C, Ar), 121.6 (CH, Ar), 120.5 (CH, Ar), 106.6 (CH, Ar), 94.3 (CH, Ar), 57.4 (C), 41.7 (CH2), 37.5 (CH2), 35.3 (CH2), 32.2 (CH2), 28.9 (CH3), 18.8 (CH3), 17.4 (CH2); ESI-LRMS m/z: 269 [M + H]+; ESI-HRMS m/z calcd for M + H+ 269.1648, found: 269.1645. The characterization data is in accordance with that reported in [45].
11-Chloro-13b-methyl-2,3,6,7-tetrahydro-1H-pyrido[2’,1’:3,4]pyrazino[1,2-a]indol-4(13bH)-one (SF10c): pale yellow oil (75.5 mg, yield 52%). 1H-NMR (300 MHz, CDCl3) δ 1.70 (s, 3H), 2.20–1.83 (m, 3H), 2.63–2.27 (m, 3H), 3.37–3.22 (m, 1H), 3.98–3.86 (m, 1H), 4.14 (dd, J = 11.6, 4.2 Hz, 1H), 5.16 (dd, J = 13.8, 4.2 Hz, 1H), 6.18 (s, 1H), 7.22–7.09 (m, 2H), 7.55–7.48 (m, 1H); 13C-NMR (100 MHz, CDCl3) δ 169.4 (CO), 143.4 (C, Ar), 134.0 (C, Ar), 129.1 (C, Ar), 126.0 (C, Ar), 121.6 (CH, Ar), 119.8 (CH, Ar), 110.0 (CH, Ar), 95.5 (CH, Ar), 57.3 (C), 41.7 (CH2), 37.4 (CH2), 35.1 (CH2), 32.1 (CH2), 28.7 (CH3), 17.3 (CH2); ESI-LRMS m/z: 291 ([M + H]+, Cl37), 289 ([M + H]+, Cl35); ESI-HRMS m/z calcd for M + H+ 289.1102, found: 289.1099. The characterization data is in accordance with that reported in [45].
13b-Methyl-6,7-dihydroisoindolo[1’,2’:3,4]pyrazino[1,2-a]indol-9(13bH)-one (SF11a): pale yellow solid (95.6 mg, yield 66%), mp 149–150 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.91 (s, 3H), 3.90–3.75 (m, 2H), 4.45–4.31 (m, 1H), 4.68–4.57 (m, 1H), 6.84 (s, 1H), 7.08–7.00 (m, 1H), 7.17–7.09 (m, 1H), 7.38 (d, J = 8.1 Hz, 1H), 7.59–7.48 (m, 2H), 7.80–7.69 (m, 2H), 8.24 (d, J = 7.6 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 166.6 (CO), 150.3 (C, Ar), 137.8 (C, Ar), 135.2 (C, Ar), 132.8 (CH, Ar), 129.9 (C, Ar), 128.7 (CH, Ar), 127.3 (C, Ar), 123.1 (2 × CH, Ar), 121.2 (CH, Ar), 120.0 (2 × CH, Ar), 109.9 (CH, Ar), 97.8 (CH, Ar), 61.4 (C), 41.1 (CH2), 34.2 (CH2), 28.2 (CH3); ESI-LRMS m/z: 289 [M + H]+; ESI-HRMS m/z calcd for M + H+ 289.1335, found: 289.1332. The characterization data is in accordance with that reported in [43].
15b-Methyl-8,9-dihydro-5H-indolo[2’,1’:3,4]pyrazino[2,1-a]isoquinolin-6(15bH)-one (SF12a): yellow solid (77.6 mg, yield 51%), mp 235–236 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.89 (s, 3H), 3.20–3.08 (m, 1H), 3.68 (d, J = 19.5 Hz, 1H), 3.81–3.72 (m, 1H), 4.10 (d, J = 19.4 Hz, 1H), 4.40–4.31 (m, 1H), 4.99–4.90 (m, 1H), 6.83 (s, 1H), 7.15–7.08 (m, 1H), 7.23–7.15 (m, 2H), 7.33–7.25 (m, 2H), 7.44–7.38 (m, 2H), 7.67 (d, J = 7.7 Hz, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 168.3 (CO), 138.6 (C, Ar), 136.9 (C, Ar), 136.5 (C, Ar), 132.1 (C, Ar), 128.0 (CH, Ar), 127.6 (CH, Ar), 127.2 (C, Ar), 126.5 (CH, Ar), 124.2 (CH, Ar), 121.4 (CH, Ar), 120.3 (CH, Ar), 119.9 (CH, Ar), 109.7 (CH, Ar), 101.4 (CH, Ar), 61.8 (C), 42.8 (CH2), 37.7 (CH2), 37.4 (CH2), 28.6 (CH3); ESI-LRMS m/z: 303 [M + H]+; ESI-HRMS m/z calcd for M + H+ 303.1492, found: 303.1487. The characterization data is in accordance with that reported in [43].
13b-Methyl-5,6,7,13b-tetrahydro-1H-pyrrolo[2’,1’:3,4][1,4]diazepino[1,2-a]indol-3(2H)-one (SF13a): colorless oil (91.6 mg, yield 72%). 1H-NMR (400 MHz, CDCl3) δ 1.72 (s, 3H), 2.14–1.88 (m, 3H), 2.53–2.42 (m, 2H), 2.80–2.70 (m, 1H), 3.17–3.04 (m, 1H), 4.20 (ddd, J = 14.8, 9.1, 2.5 Hz, 1H), 4.56–4.33 (m, 2H), 6.45 (s, 1H), 7.13–7.06 (m, 1H), 7.24–7.19 (m, 1H), 7.30 (d, J = 8.3 Hz, 1H), 7.57 (d, J = 7.8 Hz, 1H); 13C-NMR (100 MHz, CDCl3) δ 174.2 (CO), 142.5 (C, Ar), 137.7 (C, Ar), 126.9 (C, Ar), 122.0 (CH, Ar), 120.7 (CH, Ar), 119.9 (CH, Ar), 109.0 (CH, Ar), 99.6 (CH, Ar), 62.4 (C), 43.4 (CH2), 38.5 (CH2), 35.0 (CH2), 30.5 (CH2), 28.0 (CH2), 26.4 (CH3); ESI-LRMS m/z: 255 [M + H]+; ESI-HRMS m/z calcd for M + H+ 255.1492, found: 255.1489. The characterization data is in accordance with that reported in [45].
11-Bromo-13b-methyl-5,6,7,13b-tetrahydro-1H-pyrrolo[2’,1’:3,4][1,4]diazepino[1,2-a]indol-3(2H)-one (SF13b): pale yellow oil (89.3 mg, yield 54%). 1H-NMR (300 MHz, CDCl3) δ 1.71 (s, 3H), 2.15–1.91 (m, 3H), 2.51–2.40 (m, 2H), 2.77–2.66 (m, 1H), 3.16–3.03 (m, 1H), 4.18 (ddd, J = 14.8, 8.8, 2.7 Hz, 1H), 4.46–4.32 (m, 2H), 6.37 (s, 1H), 7.14 (d, J = 8.8 Hz, 1H), 7.30–7.26 (m, 1H), 7.66 (d, J = 1.9 Hz, 1H); 13C-NMR (100 MHz, CDCl3) δ 174.1 (CO), 143.7 (C, Ar), 136.4 (C, Ar), 128.5 (C, Ar), 124.8 (CH, Ar), 123.1 (CH, Ar), 112.9 (C, Ar), 110.6 (CH, Ar), 99.1 (CH, Ar), 62.3 (C), 43.7 (CH2), 38.5 (CH2), 34.9 (CH2), 30.4 (CH2), 27.9 (CH2), 26.3 (CH3); ESI-LRMS m/z: 335 ([M + H]+, Br81), 333 ([M + H]+, Br79); ESI-HRMS m/z calcd for M + H+ 333.0597, found: 333.0595. The characterization data is in accordance with that reported in [45].
12,13b-Dimethyl-5,6,7,13b-tetrahydro-1H-pyrrolo[2’,1’:3,4][1,4]diazepino[1,2-a]indol-3(2H)-one (SF13c): colorless oil (107.1 mg, yield 80%). 1H-NMR (300 MHz, CDCl3) δ 1.73 (s, 3H), 2.03–1.82 (m, 2H), 2.17–2.07 (m, 1H), 2.54–2.44 (m, 2H), 2.55 (s, 3H), 2.85–2.70 (m, 1H), 3.17–3.04 (m, 1H), 4.25–4.13 (m, 1H), 4.52–4.33 (m, 2H), 6.46 (s, 1H), 6.95–6.88 (m, 1H), 7.18–7.12 (m, 2H); 13C-NMR (100 MHz, CDCl3) δ 174.0 (CO), 141.8 (C, Ar), 137.3 (C, Ar), 130.1 (C, Ar), 126.5 (C, Ar), 122.1 (CH, Ar), 120.0 (CH, Ar), 106.6 (CH, Ar), 97.9 (CH, Ar), 62.3 (C), 43.5 (CH2), 38.4 (CH2), 34.8 (CH2), 30.4 (CH2), 28.0 (CH2), 26.3 (CH3), 18.6 (CH3); ESI-LRMS m/z: 269 [M + H]+; ESI-HRMS m/z calcd for M + H+ 269.1648, found: 269.1644. The characterization data is in accordance with that reported in [45].
14b-Methyl-1,14b-dihydroindolo[1,2-a]pyrrolo[2,1-c]quinoxalin-3(2H)-one (SF14a): pale yellow solid (123.5 mg, yield 86%), mp 179–180 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.39 (s, 3H), 2.69–2.45 (m, 3H), 2.95–2.81 (m, 1H), 6.64 (s, 1H), 7.25–7.17 (m, 1H), 7.35–7.26 (m, 2H), 7.46–7.39 (m, 1H), 7.67 (d, J = 7.7 Hz, 1H), 8.13–8.04 (m, 2H), 8.16 (d, J = 8.1 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 172.2 (CO), 142.7 (C, Ar), 133.1 (C, Ar), 129.3 (C, Ar), 128.8 (C, Ar), 125.8 (CH, Ar), 125.7 (C, Ar), 124.1 (CH, Ar), 123.0 (CH, Ar), 122.7 (CH, Ar), 121.4 (CH, Ar), 121.2 (CH, Ar), 117.2 (CH, Ar), 111.8 (CH, Ar), 97.6 (CH, Ar), 59.4 (C), 31.2 (CH2), 30.0 (CH2), 25.9 (CH3); ESI-LRMS m/z: 289 [M + H]+; ESI-HRMS m/z calcd for M + H+ 289.1335, found: 289.1331. The characterization data is in accordance with that reported in [43].
2-Hexyl-14b-methyl-1,14b-dihydroindolo[1,2-a]pyrrolo[2,1-c]quinoxalin-3(2H)-one (SF14b): pale yellow oil (159.8 mg, yield 86% (dr = 1.5:1)), and the two diastereomers were inseparable by chromatography. 1H-NMR (600 MHz, CDCl3) δ 1.00–0.80 (m, 3.02H), 1.53–1.22 (m, 11.93H), 2.09–1.96 (m, 1.03H), 2.14–2.08 (m, 0.34H), 2.35–2.27 (m, 0.53H), 2.62–2.52 (m, 0.31H), 2.81–2.73 (m, 0.53H), 2.91–2.82 (m, 0.53H), 3.03–2.94 (m, 0.32H), 6.44 (s, 0.51H), 6.47 (s, 0.34H), 7.25–7.15 (m, 1.61H), 7.36–7.27 (m, 1.91H), 7.43–7.37 (m, 0.41H), 7.68–7.61 (m, 0.92H), 7.86 (dd, J = 7.9, 1.3 Hz, 0.34H), 8.06–7.95 (m, 1.91H), 8.28 (dd, J = 8.1, 1.4 Hz, 0.53H); 13C-NMR (150 MHz, CDCl3) δ 175.3 (CO), 174.1 (CO), 143.4 (C, Ar), 141.9 (C, Ar), 134.4 (C, Ar), 133.6 (C, Ar), 130.6 (C, Ar), 129.8 (C, Ar), 129.5 (C, Ar), 129.4 (C, Ar), 126.4 (CH, Ar), 126.2 (C, Ar), 125.6 (CH, Ar), 125.4 (CH, Ar), 124.3 (2 × CH, Ar), 123.1 (2 × CH, Ar), 122.8 (CH, Ar), 121.6 (CH, Ar), 121.3 (2 × CH, Ar), 117.2 (CH, Ar), 117.1 (CH, Ar), 111.9 (CH, Ar), 111.7 (CH, Ar), 97.4 (CH, Ar), 97.1 (CH, Ar), 58.3 (C), 41.9 (CH), 41.5 (CH), 39.5 (CH2), 37.7 (CH2), 32.1 (CH2), 31.9 (CH2), 31.8 (CH2), 30.9 (CH2), 29.3 (CH2), 29.3 (CH2), 28.9 (CH3), 27.6 (CH2), 27.2 (CH2), 26.2 (CH3), 22.8 (CH2), 22.7 (CH2), 14.2 (2 × CH3); ESI-LRMS m/z: 373 [M + H]+; ESI-HRMS m/z calcd for M + H+ 373.2274, found: 373.2273.
15b-Methyl-2,3-dihydro-1H-indolo[1,2-a]pyrido[2,1-c]quinoxalin-4(15bH)-one (SF15): pale yellow oil (89.4 mg, yield 59%). 1H-NMR (400 MHz, DMSO-d6) δ 1.29 (s, 3H), 1.97–1.75 (m, 2H), 2.42–2.28 (m, 2H), 2.71–2.56 (m, 2H), 6.60 (s, 1H), 7.23–7.17 (m, 1H), 7.33–7.24 (m, 2H), 7.47–7.40 (m, 1H), 7.71–7.64 (m, 2H), 8.08–8.01 (m, 2H); 13C-NMR (100 MHz, DMSO-d6) δ 168.2 (CO), 142.5 (C, Ar), 133.2 (C, Ar), 130.5 (C, Ar), 129.1 (C, Ar), 128.4 (CH, Ar), 127.8 (C, Ar), 126.6 (CH, Ar), 123.4 (CH, Ar), 122.8 (CH, Ar), 121.2 (2 × CH, Ar), 116.9 (CH, Ar), 111.5 (CH, Ar), 96.8 (CH, Ar), 57.3 (C), 33.3 (CH2), 32.9 (CH2), 27.7 (CH3), 17.1 (CH2); ESI-LRMS m/z: 303 [M + H]+; ESI-HRMS m/z calcd for M + H+ 303.1492, found: 303.1486. The characterization data is in accordance with that reported in [43].
15b-Mmethylindolo[1,2-a]isoindolo[1,2-c]quinoxalin-11(15bH)-one (SF16): white solid (100.5 mg, yield 60%), mp 149–150 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.65 (s, 3H), 6.77 (s, 1H), 7.24–7.18 (m, 1H), 7.35–7.29 (m, 1H), 7.47–7.40 (m, 1H), 7.57–7.51 (m, 1H), 7.65 (d, J = 7.5 Hz, 1H), 7.76–7.68 (m, 1H), 7.96–7.87 (m, 2H), 8.04 (dd, J = 7.9, 1.5 Hz, 1H), 8.15 (d, J = 8.4 Hz, 1H), 8.31–8.26 (m, 1H), 8.35 (d, J = 7.6 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 164.1 (CO), 146.4 (C, Ar), 137.1 (C, Ar), 133.9 (C, Ar), 133.5 (CH, Ar), 129.7 (C, Ar), 129.5 (CH, Ar), 129.3 (C, Ar), 128.9 (C, Ar), 126.5 (CH, Ar), 124.6 (C, Ar), 124.4 (CH, Ar), 124.0 (CH, Ar), 123.9 (CH, Ar), 123.7 (CH, Ar), 123.5 (CH, Ar), 121.6 (CH, Ar), 121.3 (CH, Ar), 117.5 (CH, Ar), 111.9 (CH, Ar), 99.3 (CH, Ar), 61.2 (C), 26.3 (CH3); ESI-LRMS m/z: 337 [M + H]+; ESI-HRMS m/z calcd for M + H+ 337.1335, found: 337.1329. The characterization data is in accordance with that reported in [43].
11b-Methyl-4,5-dihydro-3H-pyrrolo[3’,2’:3,4]pyrido[2,1-a]isoindol-7(11bH)-one (SF17): pale yellow solid (77.1 mg, yield 65%), mp 237–238 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.66 (s, 3H), 2.66–2.54 (m, 2H), 3.41–3.27 (m, 1H), 4.48–4.37 (m, 1H), 6.31–6.26 (m, 1H), 6.62–6.55 (m, 1H), 7.49–7.42 (m, 1H), 7.68–7.60 (m, 2H), 7.97 (d, J = 7.6 Hz, 1H), 10.56 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 166.8 (CO), 152.2 (C, Ar), 131.9 (CH, Ar), 130.0 (C, Ar), 127.8 (CH, Ar), 122.7 (CH, Ar), 122.7 (C, Ar), 122.3 (CH, Ar), 119.9 (C, Ar), 116.8 (CH, Ar), 104.0 (CH, Ar), 62.3 (C), 34.5 (CH2), 27.7 (CH3), 22.6 (CH2); ESI-LRMS m/z: 239 [M + H]+; ESI-HRMS m/z calcd for M + H+ 239.1179, found: 239.1175. The characterization data is in accordance with that reported in [43].
12b-Methyl-5,6-dihydropyrrolo[2’,1’:3,4]pyrazino[2,1-a]isoindol-8(12bH)-one (SF18): pale yellow solid (110.9 mg, yield 93%), mp 156–157 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.76 (s, 3H), 3.69–3.60 (m, 1H), 3.78–3.70 (m, 1H), 4.08 (dd, J = 12.0, 3.6 Hz, 1H), 4.45 (dd, J = 13.3, 4.2 Hz, 1H), 6.06–5.98 (m, 1H), 6.39–6.31 (m, 1H), 6.67–6.60 (m, 1H), 7.55–7.47 (m, 1H), 7.75–7.65 (m, 2H), 8.06 (d, J = 7.9 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 166.8 (CO), 151.1 (C, Ar), 132.6 (CH, Ar), 130.0 (C, Ar), 129.6 (C, Ar), 128.4 (CH, Ar), 122.9 (CH, Ar), 122.8 (CH, Ar), 119.4 (CH, Ar), 107.8 (CH, Ar), 104.2 (CH, Ar), 61.3 (C), 43.8 (CH2), 34.9 (CH2), 28.5 (CH3); ESI-LRMS m/z: 239 [M + H]+; ESI-HRMS m/z calcd for M + H+ 239.1179, found: 239.1175. The characterization data is in accordance with that reported in [43].
9a-Methyl-4,5,9,9a-tetrahydrothieno[2,3-g]indolizin-7(8H)-one (SF19): pale yellow solid (65.1 mg, yield 63%), mp 128–129 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.42 (s, 3H), 1.90–1.79 (m, 1H), 2.31–2.15 (m, 2H), 2.61–2.52 (m, 1H), 2.74–2.62 (m, 1H), 2.85–2.76 (m, 1H), 3.09–2.98 (m, 1H), 4.15 (dd, J = 13.2, 5.6 Hz, 1H), 6.99 (d, J = 5.2 Hz, 1H), 7.37 (d, J = 5.1 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.6 (CO), 141.8 (C, Ar), 131.3 (C, Ar), 124.2 (CH, Ar), 124.1 (CH, Ar), 60.4 (C), 33.7 (CH2), 33.4 (CH2), 30.1 (CH2), 25.8 (CH3), 24.3 (CH2); ESI-LRMS m/z: 208 [M + H]+; ESI-HRMS m/z calcd for M + H+ 208.0791, found: 208.0788. The characterization data is in accordance with that reported in [43].
11b-Methyl-4,5-dihydrothieno[3’,2’:3,4]pyrido[2,1-a]isoindol-7(11bH)-one (SF20): yellow solid (105.6 mg, yield 83%), mp 199–200 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.75 (s, 3H), 2.82–2.71 (m, 1H), 2.93–2.84 (m, 1H), 3.46–3.38 (m, 1H), 4.47 (dd, J = 13.4, 5.7 Hz, 1H), 7.39 (d, J = 5.3 Hz, 1H), 7.56–7.48 (m, 2H), 7.74–7.63 (m, 2H), 8.16 (d, J = 7.6 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 166.8 (CO), 150.2 (C, Ar), 137.8 (C, Ar), 132.8 (C, Ar), 132.3 (CH, Ar), 130.1 (C, Ar), 128.4 (CH, Ar), 125.3 (CH, Ar), 124.0 (CH, Ar), 123.0 (2 × CH, Ar), 63.4 (C), 34.7 (CH2), 27.0 (CH3), 24.8 (CH2); ESI-LRMS m/z: 256 [M + H]+; ESI-HRMS m/z calcd for M + H+ 256.0791, found: 256.0785. The characterization data is in accordance with that reported in [43].
9a-Methyl-4,5,9,9a-tetrahydrothieno[3,2-g]indolizin-7(8H)-one (SF21): colorless oil (93.3 mg, yield 90%). 1H-NMR (500 MHz, DMSO-d6) δ 1.50 (s, 3H), 2.03–1.92 (m, 1H), 2.32–2.19 (m, 2H), 2.61–2.51 (m, 2H), 2.72–2.63 (m, 1H), 3.07–2.96 (m, 1H), 4.11 (dd, J = 13.3, 6.2 Hz, 1H), 6.80 (d, J = 5.1 Hz, 1H), 7.40 (d, J = 5.0 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.7 (CO), 141.8 (C, Ar), 132.3 (C, Ar), 127.1 (CH, Ar), 123.5 (CH, Ar), 60.4 (C), 35.1 (CH2), 33.6 (CH2), 30.2 (CH2), 27.9 (CH3), 25.1 (CH2); ESI-LRMS m/z: 208 [M + H]+; ESI-HRMS m/z calcd for M + H+ 208.0791, found: 208.0788. The characterization data is in accordance with that reported in [43].
11b-Methyl-4,5-dihydrothieno[2’,3’:3,4]pyrido[2,1-a]isoindol-7(11bH)-one (SF22): pale yellow solid (108.6 mg, yield 85%), mp 137–138 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.82 (s, 3H), 2.62 (ddd, J = 16.2, 11.7, 6.5 Hz, 1H), 2.76 (dd, J = 16.2, 4.5 Hz, 1H), 3.40 (ddd, J = 13.4, 12.0, 4.7 Hz, 1H), 4.43 (dd, J = 13.5, 6.2 Hz, 1H), 6.82 (d, J = 5.1 Hz, 1H), 7.45 (d, J = 5.1 Hz, 1H), 7.57–7.49 (m, 1H), 7.76–7.67 (m, 2H), 7.97–7.90 (m, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 166.5 (CO), 150.2 (C, Ar), 138.0 (C, Ar), 133.4 (C, Ar), 132.5 (CH, Ar), 130.0 (C, Ar), 128.7 (CH, Ar), 127.1 (CH, Ar), 124.4 (CH, Ar), 123.1 (CH, Ar), 122.4 (CH, Ar), 63.1 (C), 34.3 (CH2), 28.8 (CH3), 25.6 (CH2); ESI-LRMS m/z: 256 [M + H]+; ESI-HRMS m/z calcd for M + H+ 256.0791, found: 256.0789. The characterization data is in accordance with that reported in [43].
12b-Methyl-8,12b-dihydro-4H-thieno[2’,3’:3,4]pyrido[2,1-a]isoquinolin-7(5H)-one (SF23): pale yellow solid (63.4 mg, yield 47%), mp 155–156 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.87 (s, 3H), 2.50–2.44 (m, 1H), 2.76–2.68 (m, 1H), 3.10–2.99 (m, 1H), 3.64 (d, J = 20.3 Hz, 1H), 3.97 (d, J = 20.2 Hz, 1H), 4.90–4.79 (m, 1H), 6.91 (d, J = 5.1 Hz, 1H), 7.32–7.22 (m, 3H), 7.55 (d, J = 5.1 Hz, 1H), 7.77–7.71 (m, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 167.9 (CO), 139.0 (C, Ar), 138.8 (C, Ar), 136.4 (C, Ar), 131.3 (C, Ar), 128.0 (CH, Ar), 127.6 (CH, Ar), 127.4 (CH, Ar), 126.5 (CH, Ar), 124.8 (CH, Ar), 124.5 (CH, Ar), 62.6 (C), 37.0 (CH2), 36.7 (CH2), 31.7 (CH3), 25.2 (CH2); ESI-LRMS m/z: 270 [M + H]+; ESI-HRMS m/z calcd for M + H+ 270.0947, found: 270.0942. The characterization data is in accordance with that reported in [43].
8,9-Dimethoxy-10b-methyl-1,5,6,10b-tetrahydropyrrolo[2,1-a]isoquinolin-3(2H)-one (SF24a): white solid (114.5 mg, yield 88%), mp 53–54 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.45 (s, 3H), 1.93–1.82 (m, 1H), 2.25–2.15 (m, 1H), 2.45–2.37 (m, 1H), 2.59–2.47 (m, 1H), 2.70–2.62 (m, 2H), 3.05–2.96 (m, 1H), 3.71 (s, 3H), 3.75 (s, 3H), 4.08–4.00 (m, 1H), 6.66 (s, 1H), 6.78 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.3 (CO), 147.7 (C, Ar), 147.4 (C, Ar), 134.9 (C, Ar), 124.1 (C, Ar), 111.9 (CH, Ar), 108.7 (CH, Ar), 60.3 (C), 55.8 (OCH3), 55.5 (OCH3), 34.3 (CH2), 33.4 (CH2), 30.2 (CH2), 27.7 (CH2), 26.8 (CH3); ESI-LRMS m/z: 262 [M + H]+; ESI-HRMS m/z calcd for M + H+ 262.1438, found: 262.1434. The characterization data is in accordance with that reported in [43].
2-Hexyl-8,9-dimethoxy-10b-methyl-1,5,6,10b-tetrahydropyrrolo[2,1-a]isoquinolin-3(2H)-one (SF24b): white solid (81.2 mg, yield 47% (dr = 4:1)), mp 65–67 °C, and the two diastereomers were inseparable by chromatography. 1H-NMR (400 MHz, DMSO-d6) δ 0.85–0.72 (m, 3H), 1.37–0.99 (m, 10H), 1.47–1.35 (m, 3H), 1.73–1.57 (m, 1H), 2.43–2.35 (m, 1H), 2.52–2.44 (m, 1H), 2.70–2.52 (m, 2H), 3.12–2.88 (m, 1H), 3.65–3.59 (m, 3H), 3.68 (s, 3H), 4.00–3.89 (m, 1H), 6.61–6.51 (m, 1H), 6.77–6.69 (m, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 174.9 (CO), 147.5 (C, Ar), 147.4 (C, Ar), 134.7 (C, Ar), 124.4 (C, Ar), 112.0 (CH, Ar), 108.8 (CH, Ar), 59.5 (C, Ar), 55.7 (OCH3), 55.4 (OCH3), 41.2 (CH), 39.5 (CH2), 34.2 (CH2), 31.8 (CH2), 31.2 (CH2), 30.8 (CH3), 28.6 (CH2), 27.0 (CH2), 26.9 (CH2), 22.0 (CH2), 14.0 (CH3); ESI-LRMS m/z: 346 [M + H]+; ESI-HRMS m/z calcd for M + H+ 346.2377, found: 346.2375.
9,10-Dimethoxy-11b-methyl-2,3,6,7-tetrahydro-1H-pyrido[2,1-a]isoquinolin-4(11bH)-one (SF25): white solid (84.4 mg, yield 61%), mp 75–76 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.55 (s, 3H), 1.64–1.57 (m, 1H), 1.75–1.64 (m, 1H), 1.96–1.81 (m, 1H), 2.37–2.16 (m, 2H), 2.49–2.41 (m, 1H), 2.68–2.53 (m, 2H), 2.87–2.75 (m, 1H), 3.71 (s, 3H), 3.74 (s, 3H), 4.75–4.63 (m, 1H), 6.65 (s, 1H), 6.84 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 167.7 (CO), 147.4 (C, Ar), 147.2 (C, Ar), 135.2 (C, Ar), 125.5 (C, Ar), 111.7 (CH, Ar), 109.3 (CH, Ar), 58.2 (C), 55.8 (OCH3), 55.4 (OCH3), 36.7 (CH2), 34.8 (CH2), 31.5 (CH2), 28.5 (CH2), 27.3 (CH3), 16.6 (CH2); ESI-LRMS m/z: 276 [M + H]+; ESI-HRMS m/z calcd for M + H+ 276.1594, found: 276.1589. The characterization data is in accordance with that reported in [43].
2,3-Dimethoxy-12b-methyl-5,6-dihydroisoindolo[1,2-a]isoquinolin-8(12bH)-one (SF26): white solid (118.8 mg, yield 77%), mp 187–188 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.82 (s, 3H), 2.83–2.67 (m, 2H), 3.42–3.34 (m, 1H), 3.69 (s, 3H), 3.83 (s, 3H), 4.39–4.31 (m, 1H), 6.69 (s, 1H), 7.39 (s, 1H), 7.53–7.47 (m, 1H), 7.72–7.65 (m, 2H), 8.30 (d, J = 7.7 Hz, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 166.5 (CO), 151.1 (C, Ar), 147.8 (C, Ar), 147.5 (C, Ar), 132.2 (CH, Ar), 130.8 (C, Ar), 130.3 (C, Ar), 128.3 (CH, Ar), 125.3 (C, Ar), 123.4 (CH, Ar), 122.8 (CH, Ar), 112.2 (CH, Ar), 110.2 (CH, Ar), 63.4 (C), 56.1 (OCH3), 55.5 (OCH3), 34.6 (CH2), 28.7 (CH2), 28.2 (CH3); ESI-LRMS m/z: 310 [M + H]+; ESI-HRMS m/z calcd for M + H+ 310.1438, found: 310.1432. The characterization data is in accordance with that reported in [43].
3a-Methyl-2,3,3a,4-tetrahydropyrrolo[1,2-a]quinazoline-1,5-dione (SF27a): white solid (91.2 mg, yield 84%), mp 177–178 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.42 (s, 3H), 2.30–2.19 (m, 2H), 2.56–2.49 (m, 1H), 2.78–2.68 (m, 1H), 7.32–7.25 (m, 1H), 7.65–7.57 (m, 1H), 7.91 (dd, J = 7.7, 1.4 Hz, 1H), 8.07 (d, J = 8.0 Hz, 1H), 8.94 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 171.9 (CO), 161.2 (CO), 135.7 (C, Ar), 133.2 (CH, Ar), 127.7 (CH, Ar), 124.5 (CH, Ar), 119.9 (CH, Ar), 119.8 (C, Ar), 74.0 (C), 32.4 (CH2), 29.6 (CH2), 26.4 (CH3); ESI-LRMS m/z: 217 [M + H]+; ESI-HRMS m/z calcd for M + H+ 217.0972, found: 217.0969. The characterization data is in accordance with that reported in [43].
3a,4-Dimethyl-2,3,3a,4-tetrahydropyrrolo[1,2-a]quinazoline-1,5-dione (SF27b): white solid (97.5 mg, yield 85%), mp 105–107 °C. 1H-NMR (600 MHz, CDCl3) δ 1.46 (s, 3H), 2.30 (ddd, J = 12.0, 6.1, 4.0 Hz, 1H), 2.48–2.39 (m, 1H), 2.70–2.63 (m, 2H), 3.07 (s, 3H), 7.27–7.24 (m, 1H), 7.56–7.51 (m, 1H), 8.08 (dd, J = 7.8, 1.6 Hz, 1H), 8.26 (dd, J = 8.2, 0.8 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 171.3 (CO), 162.1 (CO), 135.2 (C, Ar), 133.4 (CH, Ar), 128.6 (CH, Ar), 125.0 (CH, Ar), 119.7 (CH, Ar), 119.4 (C, Ar), 78.5 (C), 32.4 (CH2), 30.3 (CH2), 27.8 (CH3), 21.8 (CH3); ESI-LRMS m/z: 231 [M + H]+; ESI-HRMS m/z calcd for M + H+ 231.1128, found: 231.1127. The characterization data is in accordance with that reported in [43].
4a-Methyl-3,4,4a,5-tetrahydro-1H-pyrido[1,2-a]quinazoline-1,6(2H)-dione (SF28a): white solid (70.7 mg, yield 61%), mp 198–199 °C. 1H-NMR (500 MHz, DMSO-d6) δ 1.35 (s, 3H), 1.90–1.74 (m, 2H), 2.09–2.02 (m, 1H), 2.17–2.09 (m, 1H), 2.48–2.39 (m, 1H), 2.62–2.54 (m, 1H), 7.36–7.28 (m, 1H), 7.60–7.52 (m, 1H), 7.66 (d, J = 8.0 Hz, 1H), 7.86 (dd, J = 7.7, 1.4 Hz, 1H), 8.85 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 168.8 (CO), 162.3 (CO), 138.1 (C, Ar), 131.8 (CH, Ar), 126.5 (CH, Ar), 126.2 (CH, Ar), 125.2 (CH, Ar), 123.7 (C, Ar), 71.1 (C), 34.7 (CH2), 33.1 (CH2), 28.3 (CH3), 16.6 (CH2); ESI-LRMS m/z: 231 [M + H]+; ESI-HRMS m/z calcd for M + H+ 231.1128, found: 231.1126. The characterization data is in accordance with that reported in [43].
4a,5-Dimethyl-3,4,4a,5-tetrahydro-1H-pyrido[1,2-a]quinazoline-1,6(2H)-dione (SF28b): white solid (78.3 mg, yield 64%), mp 138–139 °C. 1H-NMR (600 MHz, CDCl3) δ 1.39 (s, 3H), 1.98–1.86 (m, 2H), 2.17–2.10 (m, 1H), 2.44–2.36 (m, 1H), 2.63–2.55 (m, 1H), 2.74–2.65 (m, 1H), 3.12 (s, 3H), 7.32–7.28 (m, 1H), 7.54–7.49 (m, 1H), 7.62 (dd, J = 8.1, 0.9 Hz, 1H), 8.02 (dd, J = 7.8, 1.6 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 169.3 (CO), 163.6 (CO), 137.4 (C, Ar), 132.0 (CH, Ar), 127.8 (CH, Ar), 126.2 (CH, Ar), 126.0 (CH, Ar), 123.9 (C, Ar), 75.5 (C), 34.4 (CH2), 33.7 (CH2), 27.5 (CH3), 24.6 (CH3), 16.9 (CH2); ESI-LRMS m/z: 245 [M + H]+; ESI-HRMS m/z calcd for M + H+ 245.1285, found: 245.1284. The characterization data is in accordance with that reported in [43].
6a-Methyl-6,6a-dihydroisoindolo[2,1-a]quinazoline-5,11-dione (SF29a): white solid (99.3 mg, yield 75%), mp 219–220 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.72 (s, 3H), 7.42–7.36 (m, 1H), 7.70–7.64 (m, 1H), 7.77–7.71 (m, 1H), 7.85–7.78 (m, 1H), 7.89 (d, J = 7.5 Hz, 1H), 7.95 (d, J = 7.6 Hz, 1H), 8.04–7.98 (m, 2H), 9.48 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 164.4 (CO), 162.5 (CO), 146.0 (C, Ar), 135.2 (C, Ar), 133.7 (CH, Ar), 133.6 (CH, Ar), 130.1 (CH, Ar), 129.4 (C, Ar), 128.0 (CH, Ar), 125.1 (CH, Ar), 124.0 (CH, Ar), 122.9 (CH, Ar), 121.2 (CH, Ar), 119.8 (C, Ar), 74.2 (C), 27.3 (CH3); ESI-LRMS m/z: 265 [M + H]+; ESI-HRMS m/z calcd for M + H+ 265.0972, found: 265.0966. The characterization data is in accordance with that reported in [43].
6,6a-Dimethyl-6,6a-dihydroisoindolo[2,1-a]quinazoline-5,11-dione (SF29b): white solid (97.1 mg, yield 70%), mp 182–184 °C. 1H-NMR (600 MHz, CDCl3) δ 1.72 (s, 3H), 3.25 (s, 3H), 7.37–7.32 (m, 1H), 7.67–7.63 (m, 2H), 7.76–7.70 (m, 2H), 8.05–7.99 (m, 2H), 8.15 (dd, J = 8.1, 1.0 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 165.0 (CO), 163.0 (CO), 143.3 (C, Ar), 134.8 (C, Ar), 133.5 (CH, Ar), 132.8 (CH, Ar), 131.4 (C, Ar), 130.5 (CH, Ar), 128.9 (CH, Ar), 125.5 (CH, Ar), 125.2 (CH, Ar), 124.6 (CH, Ar), 121.8 (CH, Ar), 120.6 (C, Ar), 78.1 (C), 29.6 (CH3), 23.3 (CH3); ESI-LRMS m/z: 279 [M + H]+; ESI-HRMS m/z calcd for M + H+ 279.1128, found: 279.1127. The characterization data is in accordance with that reported in [43].
4b-Methyl-4bH-isoquinolino[2,1-a]quinazoline-6,12(5H,13H)-dione (SF30a): yellow solid (91.5 mg, yield 66%), mp 225–226 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.71 (s, 3H), 3.84 (d, J = 21.2 Hz, 1H), 4.16 (d, J = 21.1 Hz, 1H), 7.30–7.23 (m, 1H), 7.45–7.36 (m, 3H), 7.68–7.61 (m, 1H), 7.79–7.71 (m, 1H), 7.82 (d, J = 7.8 Hz, 1H), 7.93 (dd, J = 7.7, 1.5 Hz, 1H), 8.89 (s, 1H); 13C-NMR (100 MHz, DMSO-d6) δ 165.5 (CO), 161.9 (CO), 137.6 (C, Ar), 133.1 (C, Ar), 132.1 (CH, Ar), 128.8 (CH, Ar), 128.6 (C, Ar), 127.5 (CH, Ar), 127.2 (CH, Ar), 126.6 (CH, Ar), 126.2 (CH, Ar), 126.1 (CH, Ar), 125.7 (CH, Ar), 123.3 (C, Ar), 73.9 (C), 35.2 (CH2), 30.5 (CH3); ESI-LRMS m/z: 279 [M + H]+; ESI-HRMS m/z calcd for M + H+ 279.1128, found: 279.1122. The characterization data is in accordance with that reported in [43].
4b,5-Dimethyl-4bH-isoquinolino[2,1-a]quinazoline-6,12(5H,13H)-dione (SF30b): yellow oil (29.1 mg, yield 20%). 1H-NMR (600 MHz, CDCl3) δ 1.86 (s, 3H), 2.77 (s, 3H), 3.91 (s, 2H), 7.28 (d, J = 7.5 Hz, 1H), 7.45–7.39 (m, 2H), 7.49–7.45 (m, 1H), 7.61–7.55 (m, 2H), 7.63 (d, J = 8.0 Hz, 1H), 8.08 (dd, J = 7.9, 1.3 Hz, 1H); 13C-NMR (150 MHz, DMSO-d6) δ 169.2 (CO), 162.1 (CO), 138.6 (C, Ar), 131.9 (CH, Ar), 131.8 (C, Ar), 130.6 (C, Ar), 129.9 (CH, Ar), 128.9 (CH, Ar), 128.0 (CH, Ar), 127.7 (CH, Ar), 127.0 (CH, Ar), 126.9 (CH, Ar), 126.7 (CH, Ar), 125.2 (C, Ar), 76.7 (C), 36.2 (CH2), 29.9 (CH3), 25.7 (CH3); ESI-LRMS m/z: 293 [M + H]+; ESI-HRMS m/z calcd for M + H+ 293.1285, found: 293.1284.
3a-Methyl-2,3,3a,4-tetrahydropyrrolo[2,1-b]quinazolin-1(9H)-one (SF31): white solid (69.4 mg, yield 69%), mp 141–143 °C. 1H-NMR (600 MHz, CDCl3) δ 1.54 (s, 3H), 2.15–2.05 (m, 2H), 2.52–2.45 (m, 1H), 2.60–2.52 (m, 1H), 4.17 (d, J = 16.7 Hz, 1H), 5.02 (d, J = 16.8 Hz, 1H), 6.58 (dd, J = 8.0, 0.8 Hz, 1H), 6.81–6.76 (m, 1H), 7.07–6.99 (m, 2H); 13C-NMR (150 MHz, CDCl3) δ 174.3 (CO), 141.9 (C, Ar), 127.6 (CH, Ar), 127.0 (CH, Ar), 119.3 (CH, Ar), 117.4 (C, Ar), 116.5 (CH, Ar), 71.9 (C), 38.6 (CH2), 33.0 (CH2), 29.6 (CH2), 25.6 (CH3); ESI-LRMS m/z: 203 [M + H]+; ESI-HRMS m/z calcd for M+H+ 203.1179, found: 203.1178. The characterization data is in accordance with that reported in [43].
4b-Methyl-4b,5-dihydroisoindolo[1,2-b]quinazolin-12(10H)-one (SF32): pale yellow solid (41.1 mg, yield 33%), mp 222–223 °C. 1H-NMR (600 MHz, CDCl3) δ 1.71 (s, 3H), 4.24 (s, 1H), 4.45 (d, J = 16.9 Hz, 1H), 5.32 (d, J = 17.0 Hz, 1H), 6.69 (d, J = 8.3 Hz, 1H), 6.90–6.85 (m, 1H), 7.12–7.08 (m, 1H), 7.14 (d, J = 7.5 Hz, 1H), 7.56–7.52 (m, 1H), 7.63 (d, J = 3.9 Hz, 2H), 7.88 (d, J = 7.6 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 165.8 (CO), 147.8 (C, Ar), 140.2 (C, Ar), 132.3 (CH, Ar), 131.5 (C, Ar), 129.6 (CH, Ar), 127.9 (CH, Ar), 127.2 (CH, Ar), 124.4 (CH, Ar), 120.7 (CH, Ar), 120.5 (CH, Ar), 118.7 (C, Ar), 118.1 (CH, Ar), 71.5 (C), 38.0 (CH2), 23.9 (CH3); ESI-LRMS m/z: 251 [M + H]+; ESI-HRMS m/z calcd for M + H+ 251.1179, found: 251.1178. The characterization data is in accordance with that reported in [43].
3a-Methyl-2,3,3a,4-tetrahydro-1H-benzo[d]pyrrolo[1,2-a]imidazol-1-one (SF33): colorless oil (45.1 mg, yield 48%). 1H-NMR (600 MHz, CDCl3) δ 1.51 (s, 3H), 2.44–2.33 (m, 2H), 2.54 (ddd, J = 16.8, 8.5, 1.6 Hz, 1H), 2.78 (ddd, J = 16.8, 11.7, 8.5 Hz, 1H), 6.68 (dd, J = 7.7, 0.7 Hz, 1H), 6.84–6.79 (m, 1H), 6.98–6.93 (m, 1H), 7.43 (dd, J = 7.6, 1.1 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 173.9 (CO), 142.8 (C, Ar), 128.7 (C, Ar), 125.4 (CH, Ar), 120.3 (CH, Ar), 115.5 (CH, Ar), 110.7 (CH, Ar), 85.7 (C), 37.8 (CH2), 33.7 (CH2), 26.3 (CH3); ESI-LRMS m/z: 189 [M + H]+; ESI-HRMS m/z calcd for M + H+ 189.1022, found: 189.1023. The characterization data is in accordance with that reported in [34].
12a-Methyl-12,12a-dihydrobenzo[4,5]imidazo[2,1-a]isoquinolin-6(5H)-one (SF34): colorless oil (26.1 mg, yield 21%). 1H-NMR (600 MHz, CDCl3) δ 1.69 (s, 3H), 3.71 (d, J = 19.0 Hz, 1H), 3.88 (d, J = 19.0 Hz, 1H), 4.48 (s, 1H), 6.85 (dd, J = 7.6, 0.5 Hz, 1H), 6.93–6.89 (m, 1H), 7.02–6.97 (m, 1H), 7.24 (d, J = 7.5 Hz, 1H), 7.33–7.30 (m, 1H), 7.37–7.33 (m, 1H), 7.41 (dd, J = 7.6, 0.9 Hz, 1H), 8.02–7.98 (m, 1H);13C-NMR (150 MHz, CDCl3) δ 165.5 (CO), 139.3 (C, Ar), 139.1 (C, Ar), 131.3 (C, Ar), 129.8 (C, Ar), 128.4 (CH, Ar), 128.0 (CH, Ar), 127.6 (CH, Ar), 125.0 (CH, Ar), 123.2 (CH, Ar), 121.6 (CH, Ar), 116.6 (CH, Ar), 112.0 (CH, Ar), 82.3 (C), 38.7 (CH2), 29.5 (CH3); ESI-LRMS m/z: 251 [M + H]+; ESI-HRMS m/z calcd for M + H+ 251.1179, found: 251.1178. The characterization data is in accordance with that reported in [43].
3a-Methyl-3,3a-dihydro-1H-benzo[d]pyrrolo[2,1-b][1,3]oxazine-1,5(2H)-dione (SF35a): white solid (98.9 mg, yield 91%), mp 114–115 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.63 (s, 3H), 2.48–2.41 (m, 2H), 2.68–2.59 (m, 1H), 2.82–2.71 (m, 1H), 7.44–7.36 (m, 1H), 7.83–7.76 (m, 1H), 8.08–7.92 (m, 2H); 13C-NMR (125 MHz, DMSO-d6) δ 171.7 (CO), 161.1 (CO), 136.1 (C, Ar), 135.7 (CH, Ar), 129.9 (CH, Ar), 125.5 (CH, Ar), 120.5 (CH, Ar), 115.7 (C, Ar), 95.4 (C), 31.7 (CH2), 29.1 (CH2), 24.0 (CH3); ESI-LRMS m/z: 218 [M + H]+; ESI-HRMS m/z calcd for M + H+ 218.0812, found: 218.0809. The characterization data is in accordance with that reported in [43].
2-Hexyl-3a-methyl-3,3a-dihydro-1H-benzo[d]pyrrolo[2,1-b][1,3]oxazine-1,5(2H)-dione (SF35b): colorless oil (75.4 mg, yield 50% (dr = 5.5:1)), and the two diastereomers were inseparable by chromatography. 1H-NMR (600 MHz, DMSO-d6) for the major isomer: δ 0.91–0.84 (m, 3H), 1.42–1.21 (m, 9H), 1.65–1.61 (m, 3H), 1.85–1.78 (m, 1H), 2.13–2.07 (m, 1H), 2.71–2.62 (m, 1H), 2.91–2.83 (m, 1H), 7.41–7.37 (m, 1H), 7.81–7.78 (m, 1H), 8.00 (dd, J = 7.8, 1.5 Hz, 1H), 8.10–8.07 (m, 1H); 13C-NMR (150 MHz, DMSO-d6) for major isomer: δ 173.0 (CO), 161.0 (CO), 136.0 (CH, Ar), 135.8 (CH, Ar), 129.9 (CH, Ar), 125.3 (CH, Ar), 119.8 (CH, Ar), 115.1 (C, Ar), 93.5 (C, Ar), 39.8 (CH), 38.5 (CH2), 31.2 (CH2), 29.6 (CH2), 28.6 (CH2), 26.3 (CH2), 23.5 (CH3), 22.1 (CH2), 14.0 (CH3); ESI-LRMS m/z: 302 [M + H]+; ESI-HRMS m/z calcd for M + H+ 302.1751, found: 302.1750.
4a-Methyl-2,3,4,4a-tetrahydrobenzo[d]pyrido[2,1-b][1,3]oxazine-1,6-dione (SF36): colorless oil (88.5 mg, yield 77%). 1H-NMR (500 MHz, DMSO-d6) δ 1.55 (s, 3H), 1.86–1.77 (m, 1H), 1.95–1.86 (m, 1H), 2.31–2.21 (m, 2H), 2.55–2.48 (m, 1H), 2.68–2.58 (m, 1H), 7.46–7.39 (m, 1H), 7.77–7.70 (m, 2H), 7.98–7.93 (m, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 169.0 (CO), 161.8 (CO), 138.6 (C, Ar), 134.1 (CH, Ar), 128.7 (CH, Ar), 126.1 (2 × CH, Ar), 119.9 (C, Ar), 92.7 (C), 34.9 (CH2), 32.9 (CH2), 25.9 (CH3), 16.1 (CH2); ESI-LRMS m/z: 232 [M + H]+; ESI-HRMS m/z calcd for M + H+ 232.0968, found: 232.0965. The characterization data is in accordance with that reported in ref.43. The characterization data is in accordance with that reported in [43].
6a-Methyl-5H-benzo[4,5][1,3]oxazino[2,3-a]isoindole-5,11(6aH)-dione (SF37): pale yellow solid (83.6 mg, yield 63%), mp 138–139 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.94 (s, 3H), 7.51–7.45 (m, 1H), 7.79–7.73 (m, 1H), 7.93–7.85 (m, 2H), 7.95 (d, J = 7.5 Hz, 1H), 8.06–8.00 (m, 2H), 8.08 (dd, J = 7.8, 1.2 Hz, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 164.1 (CO), 161.3 (CO), 143.8 (C, Ar), 136.0 (CH, Ar), 135.7 (C, Ar), 134.4 (CH, Ar), 131.3 (CH, Ar), 130.2 (CH, Ar), 129.4 (C, Ar), 125.7 (CH, Ar), 124.2 (CH, Ar), 123.2 (CH, Ar), 121.2 (CH, Ar), 115.3 (C, Ar), 92.5 (C), 23.4 (CH3); ESI-LRMS m/z: 266 [M + H]+; ESI-HRMS m/z calcd for M + H+ 266.0812, found: 266.0807. The characterization data is in accordance with that reported in [43].
3a-Methyl-3,3a-dihydro-1H-naphtho[2,3-d]pyrrolo[2,1-b][1,3]oxazine-1,5(2H)-dione (SF38): yellow solid (128.1 mg, yield 96%), mp 228–230 °C. 1H-NMR (600 MHz, CDCl3) δ 1.72 (s, 3H), 2.50–2.42 (m, 1H), 2.68–2.61 (m, 1H), 2.76–2.68 (m, 1H), 2.84–2.77 (m, 1H), 7.56–7.51 (m, 1H), 7.67–7.62 (m, 1H), 7.91 (d, J = 8.3 Hz, 1H), 7.95 (d, J = 8.2 Hz, 1H), 8.44 (s, 1H), 8.72 (s, 1H); 13C-NMR (150 MHz, CDCl3) δ 171.7 (CO), 162.3 (CO), 136.7 (C, Ar), 133.2 (CH, Ar), 131.1 (C, Ar), 130.5 (C, Ar), 129.9 (CH, Ar), 129.7 (CH, Ar), 128.0 (CH, Ar), 126.9 (CH, Ar), 119.4 (CH, Ar), 115.7 (C, Ar), 95.7 (C), 32.3 (CH2), 29.6 (CH2), 25.7 (CH3); ESI-LRMS m/z: 268 [M + H]+; ESI-HRMS m/z calcd for M + H+ 268.0968, found: 268.0962. The characterization data is in accordance with that reported in [35].
4a-Methyl-2,3,4,4a-tetrahydronaphtho[2,3-d]pyrido[2,1-b][1,3]oxazine-1,6-dione (SF39): pale yellow oil (103.7 mg, yield 74%). 1H-NMR (600 MHz, CDCl3) δ 1.64 (s, 3H), 1.94–1.87 (m, 1H), 2.22–2.12 (m, 2H), 2.52–2.46 (m, 1H), 2.74–2.61 (m, 2H), 7.57–7.53 (m, 1H), 7.66–7.61 (m, 1H), 7.90 (d, J = 8.3 Hz, 1H), 7.96 (d, J = 8.2 Hz, 1H), 8.22 (s, 1H), 8.67 (s, 1H); 13C-NMR (150 MHz, CDCl3) δ 169.3 (CO), 163.3 (CO), 135.9 (C, Ar), 133.0 (C, Ar), 131.8 (CH, Ar), 130.8 (C, Ar), 129.6 (CH, Ar), 129.5 (CH, Ar), 128.2 (CH, Ar), 127.1 (CH, Ar), 124.9 (CH, Ar), 119.2 (C, Ar), 92.5 (C), 36.3 (CH2), 33.7 (CH2), 27.7 (CH3), 16.7 (CH2); ESI-LRMS m/z: 282 [M + H]+; ESI-HRMS m/z calcd for M + H+ 282.1125, found: 282.1117. The characterization data is in accordance with that reported in [43].
4b-Methyl-4bH-naphtho[2’,3’:4,5][1,3]oxazino[2,3-a]isoindole-6,14-dione (SF40): white solid (113.2 mg, yield 72%), mp 210–212 °C. 1H-NMR (600 MHz, CDCl3) δ 1.98 (s, 3H), 7.58–7.54 (m, 1H), 7.69–7.64 (m, 2H), 7.78–7.74 (m, 1H), 7.81–7.79 (m, 1H), 8.02–7.95 (m, 3H), 8.49 (s, 1H), 8.79 (s, 1H); 13C-NMR (150 MHz, CDCl3) δ 164.7 (CO), 162.4 (CO), 144.0 (C, Ar), 136.9 (C, Ar), 133.9 (CH, Ar), 133.5 (CH, Ar), 131.1 (CH, Ar), 130.9 (C, Ar), 130.4 (C, Ar), 130.3 (C, Ar), 130.0 (CH, Ar), 129.9 (CH, Ar), 128.0 (CH, Ar), 126.8 (CH, Ar), 124.8 (CH, Ar), 122.6 (CH, Ar), 119.4 (CH, Ar), 115.0 (C, Ar), 92.7 (C), 25.0 (CH3); ESI-LRMS m/z: 316 [M + H]+; ESI-HRMS m/z calcd for M + H+ 316.0968, found: 316.0959. The characterization data is in accordance with that reported in [43].
6a-Methyl-7,8-dihydro-5H-pyrido[2,3-d]pyrrolo[2,1-b][1,3]oxazine-5,9(6aH)-dione (SF41): colorless oil (65.1 mg, yield 60%). 1H-NMR (600 MHz, CDCl3) δ 1.72 (s, 3H), 2.52–2.41 (m, 1H), 2.68–2.60 (m, 1H), 2.86–2.69 (m, 2H), 7.36 (dd, J = 7.7, 4.9 Hz, 1H), 8.42 (dd, J = 7.7, 1.9 Hz, 1H), 8.81 (dd, J = 4.9, 1.9 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 171.6 (CO), 161.3 (CO), 155.1 (CH, Ar), 149.1 (C, Ar), 139.4 (CH, Ar), 122.0 (CH, Ar), 113.0 (C, Ar), 96.1 (C), 32.3 (CH2), 29.8 (CH2), 25.4 (CH3); ESI-LRMS m/z: 219 [M + H]+; ESI-HRMS m/z calcd for M + H+ 219.0764, found: 219.0763. The characterization data is in accordance with that reported in [35].
3a-Methyl-2,3,3a,5-tetrahydro-1H-benzo[d]pyrrolo[2,1-b][1,3]oxazin-1-one (SF42): pale yellow oil (33.8 mg, yield 33%). 1H-NMR (500 MHz, DMSO-d6) δ 1.45 (s, 3H), 2.07–1.97 (m, 1H), 2.29–2.19 (m, 1H), 2.43 (ddd, J = 17.1, 10.0, 2.1 Hz, 1H), 2.76–2.65 (m, 1H), 4.88 (d, J = 16.0 Hz, 1H), 4.99 (d, J = 16.0 Hz, 1H), 7.16–7.10 (m, 1H), 7.21–7.16 (m, 1H), 7.32–7.25 (m, 1H), 8.20 (d, J = 8.1 Hz, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 170.9 (CO), 132.6 (C, Ar), 127.1 (CH, Ar), 124.7 (CH, Ar), 123.7 (CH, Ar), 123.2 (C, Ar), 119.2 (CH, Ar), 89.6 (C), 62.0 (CH2), 32.5 (CH2), 29.7 (CH2), 20.7 (CH3); ESI-LRMS m/z: 204 [M + H]+; ESI-HRMS m/z calcd for M + H+ 204.1019, found: 204.1017. The characterization data is in accordance with that reported in [43].
6a-Methyl-5H-benzo[4,5][1,3]oxazino[2,3-a]isoindol-11(6aH)-one (SF43): colorless oil (22.4 mg, yield 18%). 1H-NMR (600 MHz, CDCl3) δ 1.74 (s, 3H), 4.98 (d, J = 15.3 Hz, 1H), 5.20 (d, J = 15.3 Hz, 1H), 7.14–7.10 (m, 1H), 7.19–7.15 (m, 1H), 7.40–7.35 (m, 1H), 7.59–7.54 (m, 1H), 7.68–7.60 (m, 2H), 7.91 (d, J = 7.5 Hz, 1H), 8.25 (d, J = 8.2 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 165.2 (CO), 146.1 (C, Ar), 133.1 (CH, Ar), 132.6 (C, Ar), 131.2 (C, Ar), 130.2 (CH, Ar), 127.9 (CH, Ar), 124.3 (2 × CH, Ar), 124.2 (CH, Ar), 123.0 (C, Ar), 121.8 (CH, Ar), 121.5 (CH, Ar), 88.1 (C), 63.3 (CH2), 20.8 (CH3); ESI-LRMS m/z: 252 [M + H]+; ESI-HRMS m/z calcd for M + H+ 252.1019, found: 252.1018. The characterization data is in accordance with that reported in [43].

3.4. General Procedure of the Reductive Preparation of Compounds SF47SF55

To a solution of substrates (0.3 mmol) in dry THF was added AlCl3 (0.6 mmol), then LiAlH4 (0.6 mmol) was added portionwise at 0 °C. After that, the mixture was heated to reflux for 4 h. After the reaction was cooled, the reaction mixture was diluted with dichloromethane (120.0 mL), and then water was added dropwise at 0 °C to quench the reaction under vigorous stirring conditions. The solid which precipitated out was removed by filtration, and the organic layer obtained was dried over Na2SO4. After the removal of the solvents in vacuo, the residue was purified to give SF47SF55.
11b-Methyl-2,3,5,6,11,11b-hexahydro-1H-indolizino[8,7-b]indole (SF47): pale yellow oil (40.5 mg, yield 60%). 1H-NMR (400 MHz, DMSO-d6) δ 1.67–1.54 (m, 1H), 1.80 (s, 3H), 2.04–1.93 (m, 1H), 2.20–2.08 (m, 1H), 2.94–2.85 (m, 1H), 3.07–2.96 (m, 1H), 3.30–3.22 (m, 2H), 3.56–3.47 (m, 3H), 7.05–6.99 (m, 1H), 7.15–7.09 (m, 1H), 7.35 (d, J = 8.1 Hz, 1H), 7.47 (d, J = 7.8 Hz, 1H), 11.28 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 136.3 (C, Ar), 125.7 (C, Ar), 121.8 (CH, Ar), 119.0 (CH, Ar), 118.3 (CH, Ar), 111.3 (CH, Ar), 104.5 (C, Ar), 48.9 (CH2), 42.5 (CH2), 36.4 (CH2), 24.1 (CH3), 21.0 (CH2), 15.3 (CH2); ESI-LRMS m/z: 227 [M + H]+; ESI-HRMS m/z calcd for M + H+ 227.1543, found: 227.1540.
13b-Methyl-7,8,13,13b-tetrahydro-5H-benzo[1,2]indolizino[8,7-b]indole (SF48a): pale yellow solid (52.3 mg, yield 64%), mp 204–206 °C. 1H-NMR (400 MHz, CDCl3) δ 1.95 (s, 3H), 2.62–2.53 (m, 1H), 3.28–3.13 (m, 1H), 3.54–3.38 (m, 2H), 4.27–4.18 (m, 2H), 7.13–7.03 (m, 2H), 7.21–7.13 (m, 2H), 7.31–7.22 (m, 2H), 7.85–7.74 (m, 2H), 7.93 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 135.5 (C, Ar), 131.8 (C, Ar), 126.5 (CH, Ar), 126.4 (CH, Ar), 124.9 (C, Ar), 122.8 (CH, Ar), 122.4 (CH, Ar), 120.2 (CH, Ar), 119.0 (CH, Ar), 118.3 (CH, Ar), 110.8 (CH, Ar), 52.7 (CH2), 41.0 (CH2), 26.2 (CH3), 17.1 (CH2); ESI-LRMS m/z: 275 [M + H]+; ESI-HRMS m/z calcd for M + H+ 275.1543, found: 275.1541. The characterization data is in accordance with that reported in [43].
10-Methoxy-13b-methyl-7,8,13,13b-tetrahydro-5H-benzo[1,2]indolizino[8,7-b]indole (SF48b): pale yellow oil (59.2 mg, yield 65%). 1H-NMR (400 MHz, CDCl3) δ 1.86 (s, 3H), 2.60 (ddd, J = 15.8, 4.3, 1.4 Hz, 1H), 3.13 (ddd, J = 16.2, 11.1, 6.4 Hz, 1H), 3.54–3.43 (m, 2H), 3.84 (s, 3H), 4.26–4.15 (m, 2H), 6.78 (dd, J = 8.7, 2.5 Hz, 1H), 6.93 (d, J = 2.4 Hz, 1H), 7.15 (d, J = 8.7 Hz, 1H), 7.25–7.18 (m, 2H), 7.33–7.27 (m, 1H), 7.45 (d, J = 7.4 Hz, 1H), 7.60 (s, 1H); 13C-NMR (100 MHz, CDCl3) δ 154.2 (C, Ar), 144.9 (C, Ar), 139.0 (C, Ar), 137.8 (C, Ar), 131.3 (C, Ar), 127.7 (CH, Ar), 127.7 (C, Ar), 127.4 (CH, Ar), 123.4 (CH, Ar), 121.1 (CH, Ar), 111.7 (CH, Ar), 111.7 (CH, Ar), 106.9 (C, Ar), 100.9 (CH, Ar), 65.0 (C), 56.1 (OCH3), 53.2 (CH2), 41.7 (CH2), 26.0 (CH3), 16.4 (CH2); ESI-LRMS m/z: 305 [M + H]+; ESI-HRMS m/z calcd for M + H+ 305.1648, found: 305.1645. The characterization data is in accordance with that reported in [43].
14b-Methyl-5,6,8,9,14,14b-hexahydroindolo[2’,3’:3,4]pyrido[2,1-a]isoquinoline (SF49): pale yellow oil (60.2 mg, yield 70%). 1H-NMR (400 MHz, CDCl3) δ 1.95 (s, 3H), 2.85–2.66 (m, 2H), 3.07–2.91 (m, 2H), 3.14–3.07 (m, 1H), 3.19–3.15 (m, 1H), 3.26 (ddd, J = 13.8, 6.1, 2.6 Hz, 1H), 3.76 (ddd, J = 13.9, 10.0, 5.8 Hz, 1H), 7.19–7.05 (m, 3H), 7.25–7.20 (m, 1H), 7.31–7.27 (m, 1H), 7.37–7.31 (m, 1H), 7.49 (d, J = 7.6 Hz, 1H), 7.65 (d, J = 7.8 Hz, 1H), 7.87 (s, 1H); 13C-NMR (100 MHz, CDCl3) δ140.0 (C, Ar), 137.5 (C, Ar), 135.8 (C, Ar), 134.5 (C, Ar), 130.1 (CH, Ar), 127.7 (C, Ar), 126.8 (CH, Ar), 126.6 (CH, Ar), 126.0 (CH, Ar), 121.8 (CH, Ar), 119.6 (CH, Ar), 118.5 (CH, Ar), 111.0 (CH, Ar), 105.9 (C, Ar), 58.3 (C), 46.8 (CH2), 45.5 (CH2), 30.4 (CH3), 28.8 (CH2), 17.9 (CH2); ESI-LRMS m/z: 289 [M + H]+; ESI-HRMS m/z calcd for M + H+ 289.1699, found: 289.1695. The characterization data is in accordance with that reported in [43].
11c-Methyl-2,3,5,6,7,11c-hexahydro-1H-indolizino[7,8-b]indole (SF50): pale yellow solid (62.2 mg, yield 92%), mp 249–250 °C. 1H-NMR (400 MHz, DMSO-d6) δ 1.66–1.51 (m, 1H), 1.82 (s, 3H), 2.04–1.92 (m, 1H), 2.27–2.15 (m, 1H), 2.61–2.52 (m, 1H), 3.02–2.87 (m, 1H), 3.24–3.13 (m, 1H), 3.31–3.25 (m, 1H), 3.60–3.44 (m, 3H), 7.04–6.97 (m, 1H), 7.13–7.07 (m, 1H), 7.35 (d, J = 8.0 Hz, 1H), 7.51 (d, J = 7.9 Hz, 1H), 11.24 (s, 1H); 13C-NMR (125 MHz, DMSO-d6) δ 136.2 (C, Ar), 129.5 (C, Ar), 123.3 (C, Ar), 121.3 (CH, Ar), 119.0 (CH, Ar), 118.0 (CH, Ar), 111.4 (CH, Ar), 48.7 (CH2), 41.8 (CH2), 36.1 (CH2), 24.3 (CH3), 21.2 (CH2), 16.8 (CH2); ESI-LRMS m/z: 227 [M + H]+; ESI-HRMS m/z calcd for M + H+ 227.1543, found: 227.1541.
14c-Methyl-5,6,8,9,10,14c-hexahydroindolo[3’,2’:3,4]pyrido[2,1-a]isoquinoline (SF51): pale yellow oil (63.5 mg, yield 73%). 1H-NMR (600 MHz, CDCl3) δ 2.21 (s, 3H), 2.82–2.75 (m, 1H), 2.97 (dd, J = 17.9, 6.5 Hz, 1H), 3.23–3.17 (m, 1H), 3.32–3.27 (m, 2H), 3.42–3.38 (m, 1H), 3.50–3.43 (m, 1H), 4.15–4.07 (m, 1H), 7.10–7.05 (m, 2H), 7.14–7.10 (m, 2H), 7.20–7.16 (m, 1H), 7.31 (d, J = 8.0 Hz, 1H), 7.65 (d, J = 7.9 Hz, 1H), 7.79 (d, J = 7.9 Hz, 1H), 9.64 (s, 1H); 13C-NMR (150 MHz, CDCl3) δ 142.9 (C, Ar), 136.6 (C, Ar), 132.7 (C, Ar), 131.2 (C, Ar), 129.2 (CH, Ar), 128.1 (CH, Ar), 127.3 (C, Ar), 126.3 (2 × CH, Ar), 120.8 (CH, Ar), 120.7 (CH, Ar), 119.5 (CH, Ar), 115.3 (C, Ar), 111.2 (CH, Ar), 59.8 (C), 45.8 (CH2), 45.3 (CH2), 29.9 (CH3), 23.6 (CH2), 23.1 (CH2); ESI-LRMS m/z: 289 [M + H]+; ESI-HRMS m/z calcd for M + H+ 289.1699, found: 289.1698.
15b-Methyl-6,8,9,15b-tetrahydro-5H-indolo[2’,1’:3,4]pyrazino[2,1-a]isoquinoline (SF52): pale yellow oil (34.3 mg, yield 40%). 1H-NMR (600 MHz, CDCl3) δ 1.94 (s, 3H), 2.92–2.83 (m, 1H), 3.07–2.99 (m, 1H), 3.17–3.09 (m, 1H), 3.40–3.32 (m, 1H), 3.55–3.47 (m, 1H), 3.63–3.56 (m, 1H), 4.08–4.01 (m, 1H), 4.15–4.10 (m, 1H), 6.55 (s, 1H), 7.15–7.10 (m, 2H), 7.20–7.15 (m, 3H), 7.27 (d, J = 8.0 Hz, 1H), 7.63–7.57 (m, 2H); 13C-NMR (150 MHz, CDCl3) δ 140.9 (C, Ar), 139.8 (C, Ar), 136.7 (C, Ar), 132.8 (C, Ar), 129.5 (CH, Ar), 128.0 (C, Ar), 127.8 (CH, Ar), 126.6 (CH, Ar), 126.1 (CH, Ar), 121.2 (CH, Ar), 120.3 (CH, Ar), 120.1 (CH, Ar), 109.1 (CH, Ar), 100.6 (CH, Ar), 59.2 (C), 45.7 (CH2), 45.6 (CH2), 40.4 (CH2), 31.5 (CH3), 25.3 (CH2); ESI-LRMS m/z: 289 [M + H]+; ESI-HRMS m/z calcd for M + H+ 289.1699, found: 289.1696.
11b-Methyl-4,5,7,11b-tetrahydro-3H-pyrrolo[3’,2’:3,4]pyrido[2,1-a]isoindole (SF53): pale yellow oil (57.2 mg, yield 85%). 1H-NMR (600 MHz, CDCl3) δ 1.78 (s, 3H), 3.07–2.97 (m, 1H), 3.49–3.36 (m, 2H), 3.72–3.65 (m, 1H), 4.18–4.11 (m, 1H), 4.28–4.20 (m, 1H), 6.08–6.04 (m, 1H), 6.62–6.58 (m, 1H), 7.19–7.14 (m, 2H), 7.29–7.23 (m, 1H), 7.45–7.40 (m, 1H), 7.83 (s, 1H); 13C-NMR (150 MHz, CDCl3) δ 148.1 (C, Ar), 137.8 (C, Ar), 127.2 (CH, Ar), 126.8 (CH, Ar), 122.9 (C, Ar), 122.7 (CH, Ar), 121.9 (1CH + 1C, Ar), 116.6 (CH, Ar), 105.2 (CH, Ar), 65.8 (C), 53.5 (CH2), 42.1 (CH2), 28.2 (CH3), 17.2 (CH2); ESI-LRMS m/z: 225 [M + H]+; ESI-HRMS m/z calcd for M + H+ 225.1386, found: 225.1385.
11b-Methyl-4,5,7,11b-tetrahydrothieno[3’,2’:3,4]pyrido[2,1-a]isoindole (SF54): pale yellow solid (57.9 mg, yield 80%), mp 73–74 °C. 1H-NMR (600 MHz, CDCl3) δ 1.76 (s, 3H), 2.60 (ddd, J = 16.5, 4.4, 1.5 Hz, 1H), 3.22–3.13 (m, 1H), 3.49–3.38 (m, 2H), 4.20–4.12 (m, 2H), 6.94 (d, J = 5.2 Hz, 1H), 7.00 (dd, J = 5.3, 0.7 Hz, 1H), 7.21–7.17 (m, 2H), 7.28–7.25 (m, 1H), 7.45 (d, J = 7.6 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 146.7 (C, Ar), 139.5 (C, Ar), 138.6 (C, Ar), 132.4 (C, Ar), 127.1 (2 × CH, Ar), 125.6 (CH, Ar), 122.8 (CH, Ar), 122.3 (CH, Ar), 122.1 (CH, Ar), 67.0 (C, Ar), 53.6 (CH2), 42.1 (CH2), 27.9 (CH3), 19.2 (CH2); ESI-LRMS m/z: 242 [M + H]+; ESI-HRMS m/z calcd for M + H+ 242.0998, found: 242.0997.
11b-Methyl-4,5,7,11b-tetrahydrothieno[2’,3’:3,4]pyrido[2,1-a]isoindole (SF55): yellow oil (68.6 mg, yield 95%). 1H-NMR (600 MHz, CDCl3) δ 1.85 (s, 3H), 2.47 (ddd, J = 16.4, 4.5, 1.4 Hz, 1H), 3.07–2.97 (m, 1H), 3.45–3.30 (m, 2H), 4.22–4.12 (m, 2H), 6.69 (d, J = 5.1 Hz, 1H), 7.11 (d, J = 5.0 Hz, 1H), 7.18–7.16 (m, 1H), 7.21–7.18 (m, 1H), 7.30–7.26 (m, 1H), 7.46 (d, J = 7.6 Hz, 1H); 13C-NMR (150 MHz, CDCl3) δ 147.2 (C, Ar), 142.4 (C, Ar), 138.4 (C, Ar), 132.5 (C, Ar), 127.3 (2 × CH, Ar), 126.9 (CH, Ar), 123.1 (CH, Ar), 122.8 (CH, Ar), 122.1 (CH, Ar), 67.0 (C), 53.7 (CH2), 41.8 (CH2), 29.8 (CH3), 20.2 (CH2); ESI-LRMS m/z: 242 [M + H]+; ESI-HRMS m/z calcd for M + H+ 242.0998, found: 242.0997.

4. Conclusions

In conclusion, a green and general tandem reaction between alkynoic acids and amine nucleophiles through gold catalysis in water has been developed. This process proceeds with high efficiency leading to the formation of two rings and three new bonds in a single operation. This approach features low catalyst loading, good to excellent yields, high efficiency in bond formation, high step economy, excellent selectivity, great functional group tolerance, and extraordinarily broad substrate scope, and has been successfully employed to construct a high-quality library of indole/thiophene/pyrrole/pyridine/naphthalene/benzene-fused N-heterocycles. In addition, five antimicrobial compounds were discovered from the library, suggesting the value of our strategy to identify APIs. This is the first example of the generation of pDOS compound library encompassing skeletal diversity, molecular complexity, and drug-like properties from readily available materials through gold catalysis in water. We anticipate that these valuable N-heterocycles will find more pharmaceutical applications after our further investigations.

Supplementary Materials

The following are available online. Table S1: Survey of the solvents on the yield of product SF1a, Figures S1–S12: NMR (1H-NMR, 13C-NMR, HSQC, HMBC, and 1H-1H COSY) and ESI(+)MS spectrum of SF5a, [D]n-SF5a, SF5b, [D]n-SF5b, SF1a, and [D]n-SF1a. Figure S13: preliminary screening of antibacterial activities of compounds at 100 μg/mL. Figures S14–S19: time-kill results of SF9d, SF29b, SF33, SF36, and SF41 against S. aureus strain. Figures S20–S24: CFU results of compounds SF9d, SF29b, SF33, SF36 and SF41; copies of 1H and 13C-NMR spectra of new compounds.

Author Contributions

Conceptualization, H.L. and F.Z.; Methodology, X.J. and X.L.; Formal Analysis, J.Y., J.L., and F.Z.; Investigation, X.J., P.L., and J.L.; Resources, Y.C. and X.L.; Writing—Original Draft Preparation, J.L. and F.Z.; Writing—Review and Editing, F.Z., J.W., and H.L.; Visualization, F.Z.; Supervision, H.L. and F.Z.; Project Administration, F.Z.; Funding Acquisition, J.L., H.L., and F.Z.

Funding

This research was funded by the National Natural Science Foundation of China (grants 21602022, 81620108027, and 21632008), the Major Project of Chinese National Programs for Fundamental Research and Development (grant 2015CB910304), Sichuan Science and Technology Program (grants 2018JY0345 and 2018HH007), and Chengdu Municipal Government Program of Science and Technology (grant 2016-XT00-00023-GX). The APC was funded by Chengdu University New Faculty Start-up Funding (grant 2081915037).

Acknowledgments

F.Z. gratefully acknowledges the support from the 1000 Talents Program of Sichuan Province and Chengdu Talents Program.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yarmush, M.L.; Jayaraman, A. Advances in proteomic technologies. Annu. Rev. Biomed. Eng. 2002, 4, 349–373. [Google Scholar] [CrossRef] [PubMed]
  2. Aebersold, R.; Cravatt, B.F. Proteomics–advances, applications and the challenges that remain. Trends Biotechnol. 2002, 20, S1–S2. [Google Scholar] [CrossRef]
  3. Yee, A.; Pardee, K.; Christendat, D.; Savchenko, A.; Edwards, A.M.; Arrowsmith, C.H. Structural proteomics:  Toward high-throughput structural biology as a tool in functional genomics. Acc. Chem. Res. 2003, 36, 183–189. [Google Scholar] [CrossRef] [PubMed]
  4. Cottingham, K. Government & society: New cancer center will focus on proteomics and genomics. J. Proteome Res. 2007, 6, 3369. [Google Scholar]
  5. Yates, J.R.; Osterman, A.L. Introduction:  Advances in genomics and proteomics. Chem. Rev. 2007, 107, 3363–3366. [Google Scholar]
  6. Bleicher, K.H.; Böhm, H.J.; Müller, K.; Alanine, A.I. A guide to drug discovery: Hit and lead generation: Beyond high-throughput screening. Nat. Rev. Drug Discov. 2003, 2, 369–378. [Google Scholar] [CrossRef] [PubMed]
  7. Drewry, D.H.; Macarron, R. Enhancements of screening collections to address areas of unmet medical need: An industry perspective. Curr. Opin. Chem. Biol. 2010, 14, 289–298. [Google Scholar] [CrossRef] [PubMed]
  8. Dandapani, S.; Marcaurelle, L.A. Grand challenge commentary: Accessing new chemical space for ‘undruggable’ targets. Nat. Chem. Biol. 2010, 6, 861–863. [Google Scholar] [CrossRef] [PubMed]
  9. Sauer, W.H.B.; Schwarz, M.K. Molecular shape diversity of combinatorial libraries:  A prerequisite for broad bioactivity. J. Chem. Inf. Comput. Sci. 2003, 43, 987–1003. [Google Scholar] [CrossRef] [PubMed]
  10. Haggarty, S.J. The principle of complementarity: Chemical versus biological space. Curr. Opin. Chem. Biol. 2005, 9, 296–303. [Google Scholar] [CrossRef] [PubMed]
  11. Quinn, R.J.; Carroll, A.R.; Pham, N.B.; Baron, P.; Palframan, M.E.; Suraweera, L.; Pierens, G.K.; Muresan, S. Developing a drug-like natural product library. J. Nat. Prod. 2008, 71, 464–468. [Google Scholar] [CrossRef] [PubMed]
  12. Camp, D.; Davis, R.A.; Campitelli, M.; Ebdon, J.; Quinn, R.J. Drug-like properties: Guiding principles for the design of natural product libraries. J. Nat. Prod. 2012, 75, 72–81. [Google Scholar] [CrossRef] [PubMed]
  13. Dandapani, S.; Rosse, G.; Southall, N.; Salvino, J.M.; Thomas, C.J. Selecting, acquiring, and using small molecule libraries for high-throughput screening. Curr. Protoc. Chem. Biol. 2012, 4, 177–191. [Google Scholar] [PubMed]
  14. Rzuczek, S.G.; Southern, M.R.; Disney, M.D. Studying a drug-like, RNA-focused small molecule library identifies compounds that inhibit RNA toxicity in myotonic dystrophy. ACS Chem. Biol. 2015, 10, 2706–2715. [Google Scholar] [CrossRef] [PubMed]
  15. Evans, B.E.; Rittle, K.E.; Bock, M.G.; DiPardo, R.M.; Freidinger, R.M.; Whitter, W.L.; Lundell, G.F.; Veber, D.F.; Anderson, P.S.; Chang, R.S.L.; et al. Methods for drug discovery: Development of potent, selective, orally effective cholecystokinin antagonists. J. Med. Chem. 1988, 31, 2235–2246. [Google Scholar] [CrossRef] [PubMed]
  16. Horton, D.A.; Bourne, G.T.; Smythe, M.L. The combinatorial synthesis of bicyclic privileged structures or privileged substructures. Chem. Rev. 2003, 103, 893–930. [Google Scholar] [CrossRef] [PubMed]
  17. DeSimone, R.W.; Currie, K.S.; Mitchell, S.A.; Darrow, J.W.; Pippin, D.A. Privileged structures: Applications in drug discovery. Comb. Chem. High Throughput Screen. 2004, 7, 473–494. [Google Scholar] [CrossRef] [PubMed]
  18. Welsch, M.E.; Snyder, S.A.; Stockwell, B.R. Privileged scaffolds for library design and drug discovery. Curr. Opin. Chem. Biol. 2010, 14, 347–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. González, J.F.; Ortín, I.; de la Cuesta, E.; Menéndez, J.C. Privileged scaffolds in synthesis: 2,5-piperazinediones as templates for the preparation of structurally diverse heterocycles. Chem. Soc. Rev. 2012, 41, 6902–6915. [Google Scholar] [CrossRef] [PubMed]
  20. Schreiber, S.L. Target-oriented and diversity-oriented organic synthesis in drug discovery. Science 2000, 287, 1964–1969. [Google Scholar] [CrossRef] [PubMed]
  21. Reayi, A.; Arya, P. Natural product-like chemical space: Search for chemical dissectors of macromolecular interactions. Curr. Opin. Chem. Biol. 2005, 9, 240–247. [Google Scholar] [CrossRef] [PubMed]
  22. Spandl, R.J.; Bender, A.; Spring, D.R. Diversity-oriented synthesis; a spectrum of approaches and results. Org. Biomol. Chem. 2008, 6, 1149–1158. [Google Scholar] [CrossRef] [PubMed]
  23. Oh, S.; Park, S.B. A design strategy for drug-like polyheterocycles with privileged substructures for discovery of specific small-molecule modulators. Chem. Commun. 2011, 47, 12754–12761. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, H. Construction of biologically potential library by diversity-oriented synthesis. Org. Chem. Curr. Res. 2013, 2, 1000e123. [Google Scholar] [CrossRef]
  25. Kim, H.; Tung, T.T.; Park, S.B. Privileged substructure-based diversity-oriented synthesis pathway for diverse pyrimidine-embedded polyheterocycles. Org. Lett. 2013, 15, 5814–5817. [Google Scholar] [CrossRef] [PubMed]
  26. Kim, J.; Jung, J.; Koo, J.; Cho, W.; Lee, W.S.; Kim, C.; Park, W.; Park, S.B. Diversity-oriented synthetic strategy for developing a chemical modulator of protein–protein interaction. Nat. Commun. 2016, 7, 13196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zhang, X.; Ye, D.; Sun, H.; Guo, D.; Wang, J.; Huang, H.; Zhang, X.; Jiang, H.; Liu, H. Microwave-assisted synthesis of quinazolinone derivatives by efficient and rapid iron-catalyzed cyclization in water. Green Chem. 2009, 11, 1881–1888. [Google Scholar] [CrossRef]
  28. Ye, D.; Wang, J.; Zhang, X.; Zhou, Y.; Ding, X.; Feng, E.; Sun, H.; Liu, G.; Jiang, H.; Liu, H. Gold-catalyzed intramolecular hydroamination of terminal alkynes in aqueous media: Efficient and regioselective synthesis of indole-1-carboxamides. Green Chem. 2009, 11, 1201–1208. [Google Scholar] [CrossRef]
  29. Zhou, Y.; Zhai, Y.; Li, J.; Ye, D.; Jiang, H.; Liu, H. Metal-free tandem reaction in water: An efficient and regioselective synthesis of 3-hydroxyisoindolin-1-ones. Green Chem. 2010, 12, 1397–1404. [Google Scholar] [CrossRef]
  30. Zhang, X.; Zhou, Y.; Wang, H.; Guo, D.; Ye, D.; Xu, Y.; Jiang, H.; Liu, H. Silver-catalyzed intramolecular hydroamination of alkynes in aqueous media: Efficient and regioselective synthesis for fused benzimidazoles. Green Chem. 2011, 13, 397–405. [Google Scholar] [CrossRef]
  31. Xu, S.; Zhou, Y.; Xu, J.; Jiang, H.; Liu, H. Gold-catalyzed Michael addition/intramolecular annulation cascade: An effective pathway for the chemoselective- and regioselective synthesis of tetracyclic indole derivatives in water. Green Chem. 2013, 15, 718–726. [Google Scholar] [CrossRef]
  32. Yang, T.; Campbell, L.; Dixon, D.J. A Au(I)-catalyzed N-acyl iminium ion cyclization cascade. J. Am. Chem. Soc. 2007, 129, 12070–12071. [Google Scholar] [CrossRef] [PubMed]
  33. Muratore, M.E.; Holloway, C.A.; Pilling, A.W.; Storer, R.I.; Trevitt, G.; Dixon, D.J. Enantioselective Brønsted acid-catalyzed N-acyliminium cyclization cascades. J. Am. Chem. Soc. 2009, 131, 10796–10797. [Google Scholar] [CrossRef] [PubMed]
  34. Patil, N.T.; Mutyala, A.K.; Lakshmi, P.G.V.V.; Gajula, B.; Sridhar, B.; Pottireddygari, G.R.; Rao, T.P. Au(I)-catalyzed cascade reaction involving formal double hydroamination of alkynes bearing tethered carboxylic groups: An easy access to fused dihydrobenzimidazoles and tetrahydroquinazolines. J. Org. Chem. 2010, 75, 5963–5975. [Google Scholar] [CrossRef] [PubMed]
  35. Feng, E.; Zhou, Y.; Zhang, D.; Zhang, L.; Sun, H.; Jiang, H.; Liu, H. Gold(I)-catalyzed tandem transformation: A simple approach for the synthesis of pyrrolo/pyrido[2,1-a][1,3]benzoxazinones and pyrrolo/pyrido[2,1-a]quinazolinones. J. Org. Chem. 2010, 75, 3274–3282. [Google Scholar] [CrossRef] [PubMed]
  36. Zhou, Y.; Zhai, Y.; Ji, X.; Liu, G.; Feng, E.; Ye, D.; Zhao, L.; Jiang, H.; Liu, H. Gold(I)-catalyzed one-pot tandem coupling/cyclization: An efficient synthesis of pyrrolo-/pyrido[2,1-b]benzo[d][1,3]oxazin-1-ones. Adv. Synth. Catal. 2010, 352, 373–378. [Google Scholar] [CrossRef]
  37. Zhou, Y.; Li, J.; Ji, X.; Zhou, W.; Zhang, X.; Qian, W.; Jiang, H.; Liu, H. Silver- and gold-mediated domino transformation: A strategy for synthesizing benzo[e]indolo[1,2-a]pyrrolo/pyrido[2,1-c][1,4]diazepine-3,9-diones. J. Org. Chem. 2011, 76, 1239–1249. [Google Scholar] [CrossRef] [PubMed]
  38. Patil, N.T.; Lakshmi, P.G.V.V.; Sridhar, B.; Patra, S.; Bhadra, M.P.; Patra, C.R. New linearly and angularly fused quinazolinones: Synthesis through gold(I)-catalyzed cascade reactions and anticancer activities. Eur. J. Org. Chem. 2012, 1790–1799. [Google Scholar] [CrossRef]
  39. Li, Z.; Li, J.; Yang, N.; Chen, Y.; Zhou, Y.; Ji, X.; Zhang, L.; Wang, J.; Xie, X.; Liu, H. Gold(I)-catalyzed cascade approach for the synthesis of tryptamine-based polycyclic privileged scaffolds as α1-adrenergic receptor antagonists. J. Org. Chem. 2013, 78, 10802–10811. [Google Scholar] [CrossRef] [PubMed]
  40. Ji, X.; Zhou, Y.; Wang, J.; Zhao, L.; Jiang, H.; Liu, H. Au(I)/Ag(I)-catalyzed cascade approach for the synthesis of benzo[4,5]imidazo[1,2-c]pyrrolo[1,2-a]quinazolinones. J. Org. Chem. 2013, 78, 4312–4318. [Google Scholar] [CrossRef] [PubMed]
  41. Patil, N.T.; Shinde, V.S.; Sridhar, B. Relay catalytic branching cascade: A technique to access diverse molecular scaffolds. Angew. Chem. Int. Ed. 2013, 52, 2251–2255. [Google Scholar] [CrossRef] [PubMed]
  42. Naidu, S.; Reddy, S.R. Copper-catalyzed tandem reaction in ionic liquid: An efficient reusable catalyst and solvent media for the synthesis of fused poly hetero cyclic compounds. RSC Adv. 2016, 6, 62742–62746. [Google Scholar] [CrossRef]
  43. Qiao, J.; Jia, X.; Li, P.; Liu, X.; Zhao, J.; Zhou, Y.; Wang, J.; Liu, H.; Zhao, F. Gold-catalyzed rapid construction of nitrogen-containing heterocyclic compound library with scaffold diversity and molecular complexity. Adv. Synth. Catal. 2019, 361. [Google Scholar] [CrossRef]
  44. Zhou, Y.; Ji, X.; Liu, G.; Zhang, D.; Zhao, L.; Jiang, H.; Liu, H. Gold(I)-catalyzed cascade for synthesis of pyrrolo[1,2-a:2′,1′-c]-/Pyrido[2,1-c]pyrrolo[1,2-a]quinoxalinones. Adv. Synth. Catal. 2010, 352, 1711–1717. [Google Scholar] [CrossRef]
  45. Feng, E.; Zhou, Y.; Zhao, F.; Chen, X.; Zhang, L.; Jiang, H.; Liu, H. Gold-catalyzed tandem reaction in water: An efficient and convenient synthesis of fused polycyclic indoles. Green Chem. 2012, 14, 1888–1895. [Google Scholar] [CrossRef]
  46. Aplander, K.; Hidestal, O.; Katebzadeh, K.; Lindstrom, U.M. A green and facile route to γ- and δ-lactones via efficient Pinner-cyclization of hydroxynitriles in water. Green Chem. 2006, 8, 22–24. [Google Scholar] [CrossRef]
  47. Li, C.J.; Chen, L. Organic chemistry in water. Chem. Soc. Rev. 2006, 35, 68–82. [Google Scholar] [CrossRef] [PubMed]
  48. Chanda, A.; Fokin, V.V. Organic synthesis “on water”. Chem. Rev. 2009, 109, 725–748. [Google Scholar] [CrossRef] [PubMed]
  49. Kumaravel, K.; Vasuki, G. Multi-component reactions in water. Curr. Org. Chem. 2009, 13, 1820–1841. [Google Scholar] [CrossRef]
  50. Gupta, M.; Paul, S.; Gupta, R. General aspects of 12 basic principles of green chemistry with applications. Curr. Sci. 2010, 99, 1341–1360. [Google Scholar]
  51. Butler, R.N.; Coyne, A.G. Water: Nature’s reaction enforcer—Comparative effects for organic synthesis “in-water” and “on-water”. Chem. Rev. 2010, 110, 6302–6337. [Google Scholar] [CrossRef] [PubMed]
  52. Hashmi, A.S.K. Gold-catalyzed organic reactions. Chem. Rev. 2007, 107, 3180–3211. [Google Scholar] [CrossRef] [PubMed]
  53. Corma, A.; Leyva-Pérez, A.; Sabater, M.J. Gold-catalyzed carbon−heteroatom bond-forming reactions. Chem. Rev. 2011, 111, 1657–1712. [Google Scholar] [CrossRef] [PubMed]
  54. Huang, H.; Zhou, Y.; Liu, H. Recent advances in the gold-catalyzed additions to C–C multiple bonds. Beilstein J. Org. Chem. 2011, 7, 897–936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Krause, N.; Winter, C. Gold-catalyzed nucleophilic cyclization of functionalized allenes: A powerful access to carbo- and heterocycles. Chem. Rev. 2011, 111, 1994–2009. [Google Scholar] [CrossRef] [PubMed]
  56. Dorel, R.; Echavarren, A.M. Gold(I)-catalyzed activation of alkynes for the construction of molecular complexity. Chem. Rev. 2015, 115, 9028–9072. [Google Scholar] [CrossRef] [PubMed]
  57. Gao, F.; Zhou, Y.; Liu, H. Recent advances in the synthesis of heterocycles via gold-catalyzed cascade reactions: A review. Curr. Org. Chem. 2017, 21, 1530–1566. [Google Scholar] [CrossRef]
  58. Dalpozzo, R. Strategies for the asymmetric functionalization of indoles: An update. Chem. Soc. Rev. 2015, 44, 742–778. [Google Scholar] [CrossRef] [PubMed]
  59. Lindner, A.; Claassen, V.; Hendriksen, T.W.J.; Kralt, T. Reserpine analogs; phenethylamine derivatives1. J. Med. Chem. 1963, 6, 97–101. [Google Scholar] [CrossRef] [PubMed]
  60. Daugan, A.; Grondin, P.; Ruault, C.; de Gouville, A.C.L.M.; Coste, H.; Kirilovsky, J.; Hyafil, F.; Labaudinière, R. The discovery of Tadalafil:  A novel and highly selective PDE5 inhibitor. 1: 5,6,11,11a-tetrahydro-1H-imidazo[1′,5′:1,6]pyrido[3,4-b]indole-1,3(2H)-dione analogues. J. Med. Chem. 2003, 46, 4525–4532. [Google Scholar] [CrossRef] [PubMed]
  61. Daugan, A.; Grondin, P.; Ruault, C.; de Gouville, A.C.L.M.; Coste, H.; Linget, J.M.; Kirilovsky, J.; Hyafil, F.; Labaudinière, R. The discovery of Tadalafil:  A novel and highly selective PDE5 inhibitor. 2: 2,3,6,7,12,12a-hexahydropyrazino[1′,2′:1,6]pyrido[3,4-b]indole-1,4-dione analogues. J. Med. Chem. 2003, 46, 4533–4542. [Google Scholar] [CrossRef] [PubMed]
  62. Kochanowska-Karamyan, A.J.; Hamann, M.T. Marine indole alkaloids: Potential new drug leads for the control of depression and anxiety. Chem. Rev. 2010, 110, 4489–4497. [Google Scholar] [CrossRef] [PubMed]
  63. Silvestri, R. New prospects for vinblastine analogues as anticancer agents. J. Med. Chem. 2013, 56, 625–627. [Google Scholar] [CrossRef] [PubMed]
  64. Sun, H.; Zhu, L.; Yang, H.; Qian, W.; Guo, L.; Zhou, S.; Gao, B.; Li, Z.; Zhou, Y.; Jiang, H.; et al. Asymmetric total synthesis and identification of tetrahydroprotoberberine derivatives as new antipsychotic agents possessing a dopamine D1, D2 and serotonin 5-HT1A multi-action profile. Bioorg. Med. Chem. 2013, 21, 856–868. [Google Scholar] [CrossRef] [PubMed]
  65. Guo, D.; Li, J.; Lin, H.; Zhou, Y.; Chen, Y.; Zhao, F.; Sun, H.; Zhang, D.; Li, H.; Shoichet, B.K.; et al. Design, synthesis, and biological evaluation of novel tetrahydroprotoberberine derivatives (THPBs) as selective α1A-adrenoceptor antagonists. J. Med. Chem. 2016, 59, 9489–9502. [Google Scholar] [CrossRef] [PubMed]
  66. Stempel, E.; Gaich, T. Cyclohepta[b]indoles: A privileged structure motif in natural products and drug design. Acc. Chem. Res. 2016, 49, 2390–2402. [Google Scholar] [CrossRef] [PubMed]
  67. Homer, J.A.; Sperry, J. Mushroom-derived indole alkaloids. J. Nat. Prod. 2017, 80, 2178–2187. [Google Scholar] [CrossRef] [PubMed]
  68. Klas, K.R.; Kato, H.; Frisvad, J.C.; Yu, F.; Newmister, S.A.; Fraley, A.E.; Sherman, D.H.; Tsukamoto, S.; Williams, R.M. Structural and stereochemical diversity in prenylated indole alkaloids containing the bicyclo[2.2.2]diazaoctane ring system from marine and terrestrial fungi. Nat. Prod. Rep. 2018, 35, 532–558. [Google Scholar] [CrossRef] [PubMed]
  69. Sun, H.; Huang, H.; Zhang, D.; Feng, E.; Qian, W.; Zhang, L.; Chen, K.; Liu, H. Synthesis of 4-aryl-2(5H)-furanones by gold(I)-catalyzed intramolecular annulation. Adv. Synth. Catal. 2011, 353, 1413–1419. [Google Scholar] [CrossRef]
Sample Availability: Samples of the compounds SF1–SF43, SF47–SF55 are available from the authors.
Scheme 1. Gold-catalyzed tandem reactions between alkynoic acids and amine nucleophiles. (a). Dixon’s work. (b). Our previous work. (c). This work.
Scheme 1. Gold-catalyzed tandem reactions between alkynoic acids and amine nucleophiles. (a). Dixon’s work. (b). Our previous work. (c). This work.
Molecules 24 00988 sch001
Scheme 2. Generation of scaffold diversity. a Reaction conditions: alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.005 mmol), H2O (4.0 mL), and 120 °C, 24 h. b Reaction conditions: (i) alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.01 mmol), H2O (4.0 mL), 120 °C, 20 h; (ii) TFA (0.5 mmol) was added, and then 120 °C, 4 h. c Reaction conditions: (i) alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.015 mmol), H2O (4.0 mL), 140 °C, 20 h; (ii) TFA (0.5 mmol) was added, and then 140 °C, 4 h. ND = Not detected.
Scheme 2. Generation of scaffold diversity. a Reaction conditions: alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.005 mmol), H2O (4.0 mL), and 120 °C, 24 h. b Reaction conditions: (i) alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.01 mmol), H2O (4.0 mL), 120 °C, 20 h; (ii) TFA (0.5 mmol) was added, and then 120 °C, 4 h. c Reaction conditions: (i) alkynoic acids 1 (0.6 mmol), amine nucleophiles 2 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.015 mmol), H2O (4.0 mL), 140 °C, 20 h; (ii) TFA (0.5 mmol) was added, and then 140 °C, 4 h. ND = Not detected.
Molecules 24 00988 sch002
Scheme 3. Generation of scaffold diversity. a Reactions conditions: (i) Alkynoic acids 1 (0.6 mmol), amine nucleophiles 3 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.01 mmol), H2O (4.0 mL), 120 °C, 20 h; (ii) TFA (0.5 mmol) was added, and then 120 °C, 4 h. b The reaction was carried out at 140 °C. c The reaction was carried out with 5 mol% AuPPh3Cl/AgSbF6. d The reaction was performed with 5 mol% AuPPh3Cl/AgSbF6 at 140 °C. ND = Not detected.
Scheme 3. Generation of scaffold diversity. a Reactions conditions: (i) Alkynoic acids 1 (0.6 mmol), amine nucleophiles 3 (0.5 mmol), AuPPh3Cl/AgSbF6 (0.01 mmol), H2O (4.0 mL), 120 °C, 20 h; (ii) TFA (0.5 mmol) was added, and then 120 °C, 4 h. b The reaction was carried out at 140 °C. c The reaction was carried out with 5 mol% AuPPh3Cl/AgSbF6. d The reaction was performed with 5 mol% AuPPh3Cl/AgSbF6 at 140 °C. ND = Not detected.
Molecules 24 00988 sch003
Scheme 4. Derivatization of the target compounds in the library.
Scheme 4. Derivatization of the target compounds in the library.
Molecules 24 00988 sch004
Scheme 5. Deuterium-labeling experiments.; (a) the deuterium-labeling experiment of substrates 1a and 2a in D2O; (b) the deuterium-labeling experiment of substrates 1a and 2c in D2O; (c) the deuterium-labeling experiment of substrates 1a and 2d in D2O.
Scheme 5. Deuterium-labeling experiments.; (a) the deuterium-labeling experiment of substrates 1a and 2a in D2O; (b) the deuterium-labeling experiment of substrates 1a and 2c in D2O; (c) the deuterium-labeling experiment of substrates 1a and 2d in D2O.
Molecules 24 00988 sch005
Scheme 6. Hypothetic reaction pathway 1.
Scheme 6. Hypothetic reaction pathway 1.
Molecules 24 00988 sch006
Scheme 7. Hypothetic reaction pathway 2.
Scheme 7. Hypothetic reaction pathway 2.
Molecules 24 00988 sch007
Scheme 8. Mechanistic study experiments. (a) the mechanistic experiment of substrates 1a and 2a in H2O18; (b) the mechanistic experiment of substrates D and 2a without catalyst/additive.
Scheme 8. Mechanistic study experiments. (a) the mechanistic experiment of substrates 1a and 2a in H2O18; (b) the mechanistic experiment of substrates D and 2a without catalyst/additive.
Molecules 24 00988 sch008
Scheme 9. A final proposed reaction mechanism.
Scheme 9. A final proposed reaction mechanism.
Molecules 24 00988 sch009
Table 1. Reaction condition optimization for the tandem synthesis of compound SF1a a.
Table 1. Reaction condition optimization for the tandem synthesis of compound SF1a a.
Molecules 24 00988 i001
EntryCatalyst/AdditiveSolventT (°C)Yield (%) b
1-H2O10017
2PdCl2(CH3CN)2H2O10026
3Pd(PPh3)2Cl2H2O10031
4Pd(PPh3)4H2O1000
5Pd2(dba)3H2O1000
6Cu(OAc)2H2O100trace
7Cu(OH)2H2O1005
8CuClH2O100trace
9NiOAcH2O10033
10Ni(PPh3)2ClH2O10011
11Mn(OAc)2H2O10025
12Ag2CO3H2O10051
13AgOAcH2O10057
14[RuCl2(p-cym)]2H2O10048
15CoCl2H2O10038
16Co(acac)2H2O10050
17AuPPh3ClH2O10070
18 cAuPPh3ClH2O10076
19AuPPh3ClH2O12083
20AuBr3H2O12052
21AuIH2O12046
22Au1 catalyst dH2O12081
23Au2 catalyst eH2O12078
24Au3 catalyst fH2O12080
25AuPPh3Cl/Ag2CO3H2O12085
26AuPPh3Cl/AgOAcH2O12086
27AuPPh3Cl/AgOTfH2O12089
28AuPPh3Cl/AgSbF6H2O12091
a Reaction conditions: 4-pentynoic acid 1a (0.6 mmol), tryptamine 2a (0.5 mmol), catalyst/additive (0.005 mmol), and solvent (4.0 mL). b Yield refers to isolated yield. c The reaction was performed for 48 h. d Au1 catalyst = Chloro[(1,1′-biphenyl-2-yl)di-tert-butylphosphine]gold(I). e Au2 catalyst = Chloro[1,3-bis(2,6-diisopropylphenyl)imidazol-2-ylidene]gold(I). f Au3 catalyst = (Acetonitrile)[(2-biphenyl)di-tert-butylphosphine]gold(I) hexafluoroantimonate.
Table 2. MIC90 of compounds SF9d, SF29b, SF33, SF36, and SF41 against the S. aureus strain.
Table 2. MIC90 of compounds SF9d, SF29b, SF33, SF36, and SF41 against the S. aureus strain.
CompoundMIC90 (μg/mL)
SF9d100–200
SF29b50
SF33100–200
SF3610–25
SF41100

Share and Cite

MDPI and ACS Style

Jia, X.; Li, P.; Liu, X.; Lin, J.; Chu, Y.; Yu, J.; Wang, J.; Liu, H.; Zhao, F. Green and Facile Assembly of Diverse Fused N-Heterocycles Using Gold-Catalyzed Cascade Reactions in Water. Molecules 2019, 24, 988. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules24050988

AMA Style

Jia X, Li P, Liu X, Lin J, Chu Y, Yu J, Wang J, Liu H, Zhao F. Green and Facile Assembly of Diverse Fused N-Heterocycles Using Gold-Catalyzed Cascade Reactions in Water. Molecules. 2019; 24(5):988. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules24050988

Chicago/Turabian Style

Jia, Xiuwen, Pinyi Li, Xiaoyan Liu, Jiafu Lin, Yiwen Chu, Jinhai Yu, Jiang Wang, Hong Liu, and Fei Zhao. 2019. "Green and Facile Assembly of Diverse Fused N-Heterocycles Using Gold-Catalyzed Cascade Reactions in Water" Molecules 24, no. 5: 988. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules24050988

Article Metrics

Back to TopTop