Next Article in Journal
Bioresolution Production of (2R,3S)-Ethyl-3-phenylglycidate for Chemoenzymatic Synthesis of the Taxol C-13 Side Chain by Galactomyces geotrichum ZJUTZQ200, a New Epoxide-Hydrolase-Producing Strain
Previous Article in Journal
Reduction of the Nitro Group to Amine by Hydroiodic Acid to Synthesize o-Aminophenol Derivatives as Putative Degradative Markers of Neuromelanin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Class of Promising Acaricidal Tetrahydroisoquinoline Derivatives: Synthesis, Biological Evaluation and Structure-Activity Relationships

1
College of Science, Northwest A&F University, Yangling, Shaanxi 712100, China
2
College of Life Science, Northwest A&F University, Yangling, Shaanxi 712100, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2014, 19(6), 8051-8066; https://doi.org/10.3390/molecules19068051
Submission received: 1 April 2014 / Revised: 3 June 2014 / Accepted: 11 June 2014 / Published: 16 June 2014
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
As part of our continuing research on isoquinoline acaricidal drugs, this paper reports the preparation of a series of the 2-aryl-1-cyano-1,2,3,4-tetrahydroisoquinolines with various substituents on the N-phenyl ring, their in vitro acaricidal activities against Psoroptes cuniculi, a mange mite, and discusses their SAR as well. The structures of all compounds, including 12 new ones, were elucidated by analysis of UV, IR, NMR, ESI-MS, HR-MS spectra and X-ray diffraction experiments. All target compounds showed varying degrees of activity at 0.4 mg/mL. Compound 1 showed the strongest activity, with a 50% lethal concentration value (LC50) of 0.2421 μg/mL and 50% lethal time value (LT50) of 7.79 h, comparable to the standard drug ivermectin (LC50 = 0.2474 μg/mL; LT50 = 20.9 h). The SAR showed that the substitution pattern on the N-aromatic ring exerted a significant effect on the activity. The substituents 2'-F, 3'-F, 2'-Cl, 2'-Br and 2'-CF3 remarkably enhanced the activity. Generally, for the isomers with the same substituents at different positions, the order of the activity was ortho > meta > para. It was concluded that the target compounds represent a class of novel promising candidates or lead compounds for the development of new tetrahydroisoquinoline acaricidal agents.

Graphical Abstract

1. Introduction

Mites are ectoparasites that can cause a chronic skin disease, acariasis, which occurs widely in animals and human. Psoroptes cuniculi is an animal mite that mainly infects rabbits, goats, horses and sheep [1]. It causes intense pruritus in animals, with the formation of crusts and scabs, reduction of weight gain, and even death [2,3], therefore, this mite species causes serious economic losses for the animal industry.
Therapy and control of both human scabies and animal mange have been based mainly on the use of effective drugs and chemicals. Among them, ivermectin is the most clinically effective acaricide. But as it is being used increasingly, drug-resistance of mites has developed [4,5], which often leads to drug-treatment failures, recrudescence or reinfection [6]. In addition, concerns over the environmental damage [7,8] and the toxicity of many acaricides limit their application and reduces the number of safe effective products available. These problems have prompted us to dedicate great efforts to develop new effective and safe acaricides from active natural products.
Our previous research documented that 6-alkoxydihydrosanguinarines (Figure 1), derivatives of isoquinoline alkaloid sanguinarine, possessed significant acaricidal activity in vitro against P. cuniculi, compared with the commercial acaricide ivermectin [9]. Thereafter, in order to develop more effective sanguinarine-like acaricidal drugs, we designed a class of structurally simple analogues of sanguinarine, i.e., 2-aryl-3,4-dihydroisoquinolin-2-iums (ADHIQs, Figure 1), by imitating the structural characteristics of sanguinarine, and found that most of the ADHIQs had higher activity than the 6-alkoxydihydrosanguinarines and ivermectin [10].
Figure 1. Structures of sanguinarine, 6-alkoxy dihydrosanguinarines, 2-aryl-3,4-dihydroisoquinolin-2-iums and the title compounds.
Figure 1. Structures of sanguinarine, 6-alkoxy dihydrosanguinarines, 2-aryl-3,4-dihydroisoquinolin-2-iums and the title compounds.
Molecules 19 08051 g001
However, both the 6-alkoxydihydrosanguinarines and ADHIQs are not completely satisfactory because of their drawbacks of chemical instability or high chemical reactivity [11,12,13,14,15,16], which makes the drug molecules easily loss the bioactivity under physiological conditions. Thus, it is essential to improve the bioavailability of ADHIQs to develop practical isoquinoline acaricides.
The purpose of our current research is to examine in vitro the acaricidal activity of a series of the iminium moiety-modified derivatives of ADHIQs, i.e., 1-cyano-2-aryl-1,2,3,4-tetrahydroisoquinolines (CATHIQs, Figure 1), against P. cuniculi and disclose their SAR. We expected that the stability and diversity of the target compounds as well as their structural similarity to ADHIQs could provide us with new acaricidal compounds with high stability. As far as we know, this is the first report on acaricidal activity of the target compounds.

2. Results and Discussion

2.1. Chemistry

The chemical synthesis of the target compounds is outlined in Scheme 1. According to our previously reported method [10], the intermediate 2-aryl-3,4-dihydroisoquinolin-2-ium bromides were prepared in four steps by using isochromane as a starting material. At room temperature, 24 target compounds including 12 new ones (compounds 2, 7, 8, 1018) were obtained by the reaction of sodium cyanide with the corresponding 2-aryl-3,4-dihydroisoquinolin-2-ium bromides in 64%–98% yields using 75% ethanol in water (v/v) as solvent. It was noteworthy that the reaction of 2-(2-nitrophenyl)-3,4-dihydroisoquinolin-2-ium bromide with sodium cyanide under the same condition did not give the expected compound.
Scheme 1. Synthesis of the title compounds 124.
Scheme 1. Synthesis of the title compounds 124.
Molecules 19 08051 g006
The structures of all target compounds were elucidated by spectroscopic analysis. Each of the compounds showed characteristic ion peaks at m/z [M−CN], [M+H], [M+Na] in the positive electrospray ionization MS spectrum, and a strong absorption (C≡N) in the range of 2219 to 2226 cm−1 in the IR spectrum. In the 1H- and 13C-NMR spectra, all compounds revealed singlet signals in the range of δ 5.13 to 5.60 ppm assigned to H-1 and one signal in the range of δ 51.3 to 56.2 ppm due to C-1. An exception was 17, with absorption signals at 2,361 cm−1 (C≡N) in the IR spectrum, and δH-1 5.79 and δC-1 61.4 ppm in its NMR spectra. In addition, it was noteworthy that the δH-1 signals of 18 and 21 in their 1H-NMR appeared at 5.06 ppm in the higher field and 5.74 ppm [17] in the lower field, respectively. The structures of all new compounds were further confirmed by HR-MS data.
The structure of compound 1 was further supported by a single-crystal X-ray diffraction experiment (Figure 2). Its crystallographic data revealed that the dihedral angle between two aromatic rings was 52.041(42)°. In the six-membered heterocyclic ring, C7/C6/C1/C9 was approximately coplanar, and N1 and C8 deviated from it by 0.3962 Å and 0.3792 Å, respectively. In addition, there existed a 2.32 Å intra-annular hydrogen bond between H9 and F.
Figure 2. Crystal structure of compound 1.
Figure 2. Crystal structure of compound 1.
Molecules 19 08051 g002

2.2. In Vitro Acaricidal Activity and Structure-Activity Relationships

The In vitro acaricidal activity of the compounds 124 was tested according to our previous method [10]. Ivermectin, a standard acaricide, was selected as the positive drug control. The results are shown in Figure 3.
Figure 3. The acaricidal activities of compounds 124 against P. Cuniculi at 0.4 mg/mL.
Figure 3. The acaricidal activities of compounds 124 against P. Cuniculi at 0.4 mg/mL.
Molecules 19 08051 g003
Note: IVM refers to ivermectin, a standard acaricide, as positive drug control.
All compounds showed different degrees of activity at 0.4 mg/mL. Among them, 1 displayed the highest activity, with an average mortality rate of 66.7%, slightly higher than that of ivermectin (63.3%). Compounds 2, 4, 7, 13 and 24 revealed moderate activities, with average mortality rates of 36.7% to 45.0% and the other compounds exhibited lower activities with the average mortality rates of 6.7% to 30.0%.
Comparison of the activity of 24 without substituents on the N-benzene ring with that of 123 showed that the characteristics and position of the substituent on the N-phenyl ring had a significant effect on the activity. The general trend was that the introduction of 2'-F (1), 3'-F (2), 2'-Cl (4), 2'-Br (7) or 2'-CF3 (13) on the N-aromatic ring led to an obvious improvement of the activity. On the contrary, the introduction of an iodine atom (compounds 1012), methyl (compounds 1820), methoxy (compounds 2123) or nitro group (compounds 1617) to any position of N-aromatic ring caused the reduction of the activity to varying degrees.
For various isomers with the same substitution, it was clearly observed that the position of the substituent had a significant effect on their activity. Generally, except for methoxy- and nitro-substituted compounds (compounds 1617, 2123), the order of the activity of each set of isomers was 2'-substituted isomer > 3'-substituted isomer > 4'-substituted isomer. For example, the mortality rates of 2'-, 3'- and 4'-F isomers 13 were 66.7%, 44.3% and 30.0%, respectively. Therefore, for most of the substituents, the 2'-position was considered as an optimal modifiable site for the improvement of the activity.
Furthermore, comparing the activities of the compounds with various substituents at the same position, it was found that the impact of various substituents on the activity was totally different. In most cases, the intensity order of the substituents’ influence on the activity was as follows: fluorine atom (compounds 13) > chlorine atom (compounds 46) ≈ bromine atom (compounds 79) ≈ trifluoromethyl (compounds 1315) > iodine atom (compounds 1012) ≈ methyl (compounds 1820) > methoxy (compounds 2123) ≈ nitro (compounds 1617).
To explore in more detail the acaricidal potency of all compounds, compound 1 with the highest activity was subjected to an acaricidal toxicity assay using ivermectin as a standard drug control. The acaricidal activities against P. cuniculi caused by the treatment with various concentrations of 1 and ivermectin at 24 h post-treatment are shown in Figure 4. Their toxicity regression equations for concentration effects are listed in Table 1.
Figure 4. The acaricidal activities of 1 and ivermectin at various concentrations at 24 h.
Figure 4. The acaricidal activities of 1 and ivermectin at various concentrations at 24 h.
Molecules 19 08051 g004
Table 1. Toxicity regression equations for concentration effect and LC50 values (mg/L) of 1 against P. cuniculi (24 h).
Table 1. Toxicity regression equations for concentration effect and LC50 values (mg/L) of 1 against P. cuniculi (24 h).
CompoundToxicity Equation aR2LC50/mg/mL (mM)95%CI of LC50 bLinear Scope/μg/mL
1y = 2.1170x − 0.04690.99470.2421 (0.960)0.2354–0.24900.1000–1.200
Ivermectiny = 1.3165x + 1.84910.98040.2474 (0.283)0.1979–0.31020.0500–1.600
a y: The probability of the mortality; x: log [concentration (μg/mL)]; b 95% CI: lower and upper values of the confidence interval of LC50 (mg/mL) at 95% probability.
The results of Figure 4 proved that the activities of 1 and ivermectin were enhanced with the increase of their concentration. It was worth noting that there was a cross point between the two curves at a concentration of ca. 0.2 mg/mL, indicating that 1 was more effective than ivermectin at more than ca. 0.2 mg/mL. A similar case was also observed for 6-methoxydihydrosanguinarine [9]. Statistical analysis showed that both 1 and ivermectin had a significant linear correlation between the probability of mortality rates and log[concentration] values (R2 > 0.98) (Table 1). Although 1 and ivermectin showed approximately identical median lethal concentration values (LD50 = 0.2421, 0.2474 mg/mL), the toxicity regression equation of 1 had a much larger slope ratio value (k = 2.1170) than that of ivermectin (k = 1.3165), indicating that the activity of 1 had more sensitive concentration-dependence than that of ivermectin.
The acaricidal activities caused by treatment of the same concentration of 1 and ivermectin (3.0 mg/mL) at various treatment times are shown in Figure 5 and their toxicity regression equations for time effect are listed in Table 2. Like in the case of the concentration effect, the acaricidal activities of the two compounds were heightened with the prolongation of the treatment time (Figure 5).
Figure 5. The acaricidal activities of 1 and ivermectin at 3.0 mg/mL at various post-treatment times.
Figure 5. The acaricidal activities of 1 and ivermectin at 3.0 mg/mL at various post-treatment times.
Molecules 19 08051 g005
Table 2. Toxicity regression equations for treatment time effect and LT50 values of 1 against P. cuniculi (3.0 mg/mL).
Table 2. Toxicity regression equations for treatment time effect and LT50 values of 1 against P. cuniculi (3.0 mg/mL).
CompoundToxicity Equation aR2LT50/h95%CI b of LT50Linear Scope/h
1y = 5.424x + 0.16570.97427.797.59–8.035.5–14.0
Ivermectiny = 7.3816x – 4.74270.949120.8920.75–20.9317.0–23.0
a y: The probability of the inhibition rate; x: log [treat time (h)]; b 95% CI: lower and upper values of the confidence interval of LT50 (h) at 95% probability.
Meanwhile, we also observed some distinct differences between the two curves. At every treatment timepoint, compound 1 nearly showed higher activity than ivermectin. After 4 h post-treatment, the mortality rates caused by 1 began to quickly increase as the time went on, and finally reached 100% at 17 h. Unlike the case of 1, the activity of ivermectin showed one slow increase in the 6 h to 17 h range. Before 17 h post-treatment, ivermectin gave lower mite mortality rates (<30%). Even by 24 h post-treatment, ivermectin did not cause the death of all mites. Statistical analysis showed that 1 had a significant linear correlation between the mortality rate probability and log[treatment time] values (R2 = 0.9742) (Table 2) in the range of 5.5 h to 14 h, and its median lethal times (LT50 = 7.79 h) was much smaller than that of ivermectin (LT50 = 20.89 h) (Table 2). The above results showed that 1 possessed a much faster acaricidal action than ivermectin.
In our previous study, 6-alkoxydihydrosanguinarines were proven to be prodrugs of sanguinarine for the acaricidal activity [9]. 6-Alkoxydihydrosanguinarines, which structurally contains a N,O-acetal moiety, are very easily converted to the corresponding iminium ion, (i.e., sanguinarine) under acidic conditions [18] including the acidic medium of the lysosome of cells [19,20]. Like the 6-alkoxy-dihydrosanguinarines, 1-cyano-2-aryl-1,2,3,4-tetrahydroisoquinolines (CATHIQs) also have hydrolyzable properties under acidic condition (pH < 6.0). We observed that dissolving compounds 1, 2, or 3 in a buffer solution (pH = 4.3) at room temperature for 25 min resulted in an equilibrium between the compound and its corresponding iminium form (Scheme 2).
Scheme 2. Transformation between 1, 2 or 3 and its corresponding iminium form.
Scheme 2. Transformation between 1, 2 or 3 and its corresponding iminium form.
Molecules 19 08051 g007
At that time, the molar ratio of 1 to its corresponding iminium form was about 3:2 (unpublished data). In view of the structural similarity of CATHIQs, we speculated that similar hydrolysable properties should exist in the other CATHIQs. Considering the acidic conditions (pH = 3.5–5.5) in the lysosome of cells, the above transformation of CATHIQs perhaps also occurs in cells. The above analysis suggests that CATHIQs may be prodrugs or precursor drugs of their corresponding iminium salts (ADHIQs). This will be further investigated in detail in our future work. The activity of each CATHIQ listed in Figure 3 is lower than that of its corresponding iminium salt (ADHIQ) [10], which may be related with that various CATHIQs have different hydrolysable properties under acidic conditions, such as hydrolysis rates and equilibrium constants. In fact, we indeed observed that compounds 1, 2 and 3 had different susceptibility to the same acidic solutions in the range of pH 2.2–6.3.
As potential drugs, CATHIQs have higher compatibility with physiological conditions than the corresponding ADHIQs because we have proven that they have an ability to resist some biological reducing agents such as NAD(P)H and nucleophiles with amino or mercapto groups. Based on the considerations above, the present research strongly suggests that the target compounds represent a class of novel promising candidates or lead compounds for the development of new tetrahydroisoquinoline acaricidal agents. At present, in vivo testing of the CATHIQs is underway in our lab, and the corresponding results will be reported later.

3. Experimental Section

3.1. Materials and Apparatus

Ivermectin (C48H74O14, molecular weight: 875.09276, ≥91%, 22,23-dihydroavermectin B1 consisting of 95% avermectin B1a and 5% avermectin B1b) was purchased from Sigma-Aldrich Trading Co. Ltd. (Shanghai, China). Other chemicals used in the present study were purchased from J&K Chemical Ltd. (Beijing, China) and used without further purification. The intermediate 2-aryl-3,4-dihydroisoquinolin-2-ium bromides (ADHIQs in Figure 1) were prepared according to our previously reported method [10]. Melting points (mp) were determined on an XT-4 micro-melting point apparatus and are uncorrected. 1H-NMR and 13C-NMR spectra were recorded with a Bruker AVANCE III instrument operating at 500 and 125 MHz, respectively, and using TMS as an internal standard. ESI-MS were measured on a Trace mass spectrometer.

3.2. General Procedure for the Synthesis of Compounds 124

To the solution of 2-aryl-3,4-dihydroisoquinolin-2-ium bromine (0.5 mmol) in ethanol or 95% ethanol in water (ca. 20 mL), KCN (76 mg, 1.17 mmol) was added. The reaction solution was stirred at room temperature or 0 °C for 30 min, and then diluted with CHCl3 or CH2Cl2 (80 mL). The resulting solution was washed with water (3 × 30 mL), and dried over anhydrous sodium sulfate. The solvent was removed under a reduced pressure to provide crude product as an oil or solid. The crude product was subjected to column chromatography over silica gel eluting with the mixture of petroleum ether and ethyl acetate (10:1) to afford 124 as solids or liquids.
2-(2-Fluorophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (1): white solid; yield: 74%; m.p. 88–89 °C; IR (KBr) υmax cm−1: 2224 (w, C≡N), 1142 (s, C−N), 1228 (s, C−F); UV (MeOH): λmax (lg ε) 237(4.09) nm; 1H-NMR (CDCl3) δ: 7.29–7.33 (1H, m), 7.25–7.27 (2H, m), 7.21–7.23 (2H, m), 7.16–7.19 (1H, m), 7.10–7.12 (2H, m), 5.48 (1H, s, H-1), 3.60–3.51 (2H, m, H-3), 3.22 (1H, ddd, J = 17.0, 11.0, 7.0 Hz, H-4a), 2.93 (1H, d-like, H-4b); 13C-NMR (CDCl3) δ: 156.1 (d, J = 245.0 Hz, C-2'), 136.9 (d, J = 9.2 Hz, C-1'), 134.0, 129.6, 129.3, 128.7, 127.1, 126.8, 125.0 (d, J = 4.4 Hz), 121.5 (d, J = 2.0 Hz), 117.5 (C≡N), 116.4 (d, J = 20.2 Hz), 53.9 (C-1), 44.8 (C-3), 28.6 (C-4); positive ESI-MS m/z: 226 [M−CN]+, 253 [M+H]+, 275 [M+Na]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
Crystallographic data and X-ray structure analysis of 1. A colorless crystal (0.50 × 0.28 × 0.24) of 1 was grown by slow evaporation in petroleum ether-AcOEt solution. Diffraction intensity data were acquired with a CCD diffractometer with graphite-monochromated Mo radiation (λ = 0.71073 Å). Crystal data for 1: C16H13FN2, Mr = 252.28, Orthorhombic, space group Pbca, Z = 8, a = 7.4686(7), b = 14.9034(14), c = 23.239(2) Å, α = 90.00°, β = 90.00°, γ = 90.00°, V = 2586.7(4) Å3, T = 296(2) K, μ (Mo Ká) = 0.087 mm−1, 17836 reflections measured, 2402 independent reflections (Rint = 0.0252), S = 0.993. The final R1 values were 0.0363 and wR2 = 0.1182 (I > 2σ (I)). The final R1 values were 0.0472 and wR2 = 0.1324 (for all data). Crystallographic data for 1 have been deposited with the Cambridge Crystallographic Data Centre (deposition number CCDC 986701). Copies of the data can be obtained, free of charge, on application to the Director, CCDC, 12 Union Road, Cambridge CB21EZ, UK (fax: +44(0)-1223–336033 or e-mail: [email protected]. ac.uk).
2-(3-Fluorophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (2): white solid; yield: 83%; m.p. 94–95 °C; IR (KBr) υmax cm−1: 2219 (w, C≡N, 1146 (s, C−N), 1174 (s, C−F); UV (MeOH): λmax (lg ε) 246 (4.12) nm; 1H-NMR (CDCl3) δ: 7.28–7.37 (5H, m), 6.85 (1H, dd, J = 8.5, 2.5 Hz), 6.79 (1H, dt, J = 11.5, 2.5 Hz), 6.73 (1H, td, J = 8.5, 2.5 Hz), 5.54 (1H, s, H-1), 3.78–3.83 (1H, m, H-3a), 3.53 (1H, ddd, J = 12.5, 10.5, 4.0 Hz, H-3b), 3.19 (1H, ddd, J = 16.5, 10.5, 6.0 Hz, H-4a), 3.05 (1H, dt, J = 16.0, 4.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 163.8 (d, J = 243.6 Hz, C-3'), 149.8 (d, J = 9.6 Hz, C-1'), 134.6, 130.8 (d, J = 9.9 Hz), 129.3, 129.0, 127.1, 117.5 (C≡N), 112.2 (d, J = 2.6 Hz), 108.1 (d, J = 21.1 Hz), 104.1 (d, J = 25.1 Hz), 52.3 (C-1), 44.1 (C-3), 28.4 (C-4); positive ESI-MS m/z: 226 [M−CN]+, 253 [M+H]+, 275 [M+Na]+. HR-MS [M+Na]+: Calcd. for C16H13FN2Na+, 275.0960, found 275.0969.
2-(4-Fluorophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (3): white solid; yield: 88%; m.p. 124–125 °C; IR (KBr) υmax cm−1: 2226 (w, C≡N), 1142 (s, C−N), 1244 (s, C−F); UV (MeOH): λmax (lg ε) 240 (3.97) nm; 1H-NMR (CDCl3) δ: 7.23–7.34 (4H, m), 7.08 (2H, s), 7.07 (2H, d, J = 2.5 Hz), 5.40 (1H, s, H-1), 3.63 (1H, q-like, H-3a), 3.46 (1H, td, J = 11.0, 4.0 Hz, H-3b), 3.17 (1H, ddd, J = 17.0, 11.0, 6.5 Hz, H-4a), 2.96 (1H, dt, J = 16.0, 3.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 158.6 (d, J = 240.4 Hz, C-4'), 145.1 (d, J = 2.5 Hz, C-1'), 134.3, 129.5, 129.4, 128.8, 127.1, 126.9, 120.5 (d, J = 10.5 Hz), 117.4 (C≡N), 116.2 (d, J = 22.4 Hz), 54.8 (C-1), 44.8 (C-3), 28.6 (C-4); positive ESI-MS m/z: 226 [M−CN]+, 253 [M+H]+, 275 [M+Na]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-(2-Chlorophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (4): white sheet-like crystals; yield: 87%; m.p. 115–116 °C; IR (KBr) υmax cm−1: 2224 (w, C≡N), 1142 (s, C−N); UV (MeOH): λmax (lg ε) 243 (3.92) nm; 1H-NMR (CDCl3) δ: 7.44 (1H, d-like, J = 8.0 Hz), 7.30–7.35 (3H, m), 7.23–7.27 (3H, m), 7.13–7.16 (1H, m), 5.53 (1H, s, H-1), 3.62 (1H, td, J = 12.0, 4.0 Hz, H-3a), 3.46 (1H, dd, J = 12.0, 6.5 Hz, H-3b), 3.26 (1H, ddd, J = 16.5, 11.5, 6.0 Hz, H-4a), 2.93 (1H, dd, J = 16.5, 2.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 145.9 (C-1'), 134.1, 130.7, 129.7, 128.7, 128.2, 127.1, 126.7, 126.1, 123.2, 117.4 (C≡N), 53.9 (C-1), 45.6 (C-3), 28.8 (C-4); positive ESI-MS m/z: 242 [M−CN]+, 269 [M+H]+, 291[M+Na]+. The IR, 1H-NMR and 13C-NMR are in agreement with the literature data [17].
2-(3-Chlorophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (5): white powder; yield: 78%; m.p. 83–85 °C; IR (KBr) υmax cm−1: 2223 (w, C≡N), 1153 (s, C−N); UV (MeOH): λmax (lg ε) 251 (4.41) nm; 1H-NMR (CDCl3) δ: 7.23–7.32 (5H, m), 7.03 (1H, t-like), 6.92–6.97 (2H, m), 5.49 (1H, s, H-1), 3.72–3.76 (1H, m, H-3a), 3.48 (1H, ddd, J = 12.5, 10.5, 4.5 Hz, H-3b), 3.12 (1H, ddd, J = 16.0, 10.0, 5.5 Hz, H-4a), 2.97 (1H, dt, J = 16.5, 4.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 149.4 (C-1'), 135.4, 134.5, 130.6, 129.3, 129.2, 129.0, 127.1, 121.5, 117.5 (C≡N), 117.2, 115.0, 52.4 (C-1), 44.1 (C-3), 28.4 (C-4); positive ESI-MS m/z: 242[M−CN]+, 269.0[M+H]+, 291[M+Na]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-(4-Chlorophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (6): white powder; yield: 78%; m.p. 152–153 °C; IR (KBr) υmax cm−1: 2223 (w, C≡N), 1142 (s, C−N); UV (MeOH): λmax (lg ε) 253 (4.59) nm; 1H-NMR (CDCl3) δ: 7.31–7.35 (5H, m), 7.27–7.28 (1H, m), 7.02–7.05 (2H, m), 5.49 (1H, s, H-1), 3.74 (1H, ddd-like, H-3a), 3.50 (1H, ddd-like, H-3b), 3.18 (1H, ddd, J = 16.0, 10.5, 6.0 H-4a), 3.00 (1H, dt, J = 16.0, 3.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 147.0 (C-1'), 134.4, 129.6, 129.4, 129.3, 128.9, 127.1, 127.0, 118.9, 117.5 (C≡N), 53.1 (C-1), 44.3 (C-3), 28.5 (C-4); positive ESI-MS m/z: 242 [M−CN]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-(2-Bromophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (7): white powder; yield: 90%; m.p. 119–120 °C; IR (KBr) υmax cm−1: 2224 (w, C≡N); 1141 (s, C−N); UV (MeOH): λmax (lg ε) 240 (4.12) nm; 1H-NMR (CDCl3) δ: 7.62 (1H, dd, J = 7.5, 1.0 Hz), 7.40 (1H, td-like), 7.30–7.35 (2H, m), 7.23–7.27 (3H, m), 5.53 (1H, s, H-1), 3.64 (1H, dt, J = 12.0, 4.0 Hz, H-3a), 3.42 (1H, dd, J = 12.0, 6.5 Hz, H-3b), 3.27 (1H, ddd, J = 17.0, 11.5, 6.5 Hz, H-4a), 2.92 (1H, dd, J = 16.5, 3.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 147.2 (C-1'), 134.2, 133.8, 129.7, 129.5, 128.9, 128.7, 127.1, 126.7, 123.8, 120.5, 117.3(C≡N), 54.3 (C-1), 45.8 (C-3), 28.9 (C-4); positive ESI-MS m/z: 286 [M−CN]+, 313 [M+H]+. HR-MS [M+Na]+: Calcd. for C16H13BrN2Na+, 335.0160, found 335.0155.
2-(3-Bromophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (8): white sheet crystal; yield: 75%; m.p. 92–93 °C; IR (KBr) υmax cm−1: 2220 (w, C≡N), 1150 (s, C−N); UV (MeOH): λmax (lg ε) 252 (4.13) nm; 1H-NMR (CDCl3) δ: 7.31–7.37 (3H, m), 7.24–7.29 (3H, m), 7.15–7.17 (1H, m), 7.02–7.03 (1H, m), 5.53 (1H, s, H-1), 3.77–3.81 (1H, m, H-3a), 3.53 (1H, td, J = 11.5, 4.0 Hz, H-3b), 3.18 (1H, ddd, J = 16.0, 10.5, 6.0 Hz, H-4a), 3.02 (1H, d, J = 16.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 149.5 (C-1'), 134.5, 130.9, 129.4, 129.2, 129.0, 127.1, 127.0, 124.4, 123.6, 120.1, 117.5 (C≡N), 115.4, 52.4 (C-1), 44.1 (C-3), 28.4 (C-4); positive ESI-MS m/z: 286[M−CN]+. HR-MS [M+Na]+: Calcd. for C16H13BrN2Na+, 335.0160, found 335.0154.
2-(4-Bromophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (9): white sheet crystal; yield: 72%; m.p. 156–157 °C; IR (KBr) υmax cm−1: 2222 (w, C≡N), 1138 (s, C−N); UV (MeOH): λmax (lg ε) 204 (4.70), 255 (4.48) nm; 1H-NMR (CDCl3) δ: 7.45 (2H, d-like, J = 8.5 Hz), 7.28–7.34 (3H, m), 7.23–7.25 (1H, m), 6.95 (2H, d-like, J = 9.0 Hz), 5.45 (1H, s, H-1), 3.69–3.74 (1H, m, H-3a), 3.46 (1H, ddd, J = 12.5, 10.5, 4.0 Hz, H-3b), 3.17 (1H, ddd, J = 16.5, 10.5, 6.0 Hz, H-4a), 2.98 (1H, dt, J = 13.0, 3.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 147.4 (C-1'), 134.4, 132.5, 129.4, 129.2, 129.0, 127.1, 127.0, 119.1, 117.5 (C≡N), 114.4, 52.9 (C-1), 44.3 (C-3), 28.4 (C-4); positive ESI-MS m/z: 286 [M−CN]+, 313 [M+H]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-(2-Iodophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (10): white powder; yield: 83%; m.p. 127–128 °C; IR (KBr) υmax cm−1: 2223 (w, C≡N), 1138 (s, C−N); UV (MeOH): λmax (lg ε) 204 (4.01) nm; 1H-NMR (CDCl3) δ: 7.90 (1H, d, J = 7.5 Hz), 7.42–7.45 (1H, t-like, J = 7.5 Hz), 7.31–7.34 (2H, m), 7.23–7.27 (3H, m), 6.95 (1H, t, J = 7.5 Hz, H-6'), 5.43 (1H, s, H-1), 3.63–3.68 (1H, t-like, J = 11.5 Hz, H-3a), 3.35–3.38 (1H, m, H-3b), 3.23–3.30 (1H, m, H-4a), 2.92 (1H, d, J = 16.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 150.0 (C-1'), 140.1, 134.3, 129.8, 129.7, 129.5, 128.7, 127.6, 127.1, 126.7, 123.9, 117.3 (C≡N), 98.5 (C-2'), 55.1 (C-1), 46.2 (C-3), 29.1 (C-4); positive ESI-MS m/z: 334 [M−CN]+, 361 [M+H]+, 383 [M+Na]+. HR-MS [M+Na]+: Calcd. for C16H13IN2Na+, 383.0021, found 383.0015.
2-(3-Iodophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (11): white powder; yield: 79%; m.p. 124–125 °C; IR (KBr) υmax cm−1: 2220 (w, C≡N), 1135 (s, C−N); UV (MeOH): λmax (lg ε) 252 (4.30) nm; 1H-NMR (CDCl3) δ: 7.39 (1H, s, H-2’), 7.29–7.33 (4H, m), 7.24–7.25 (1H, m), 7.01–7.08 (2H, m), 5.47 (1H, s, H-1), 3.72–3.76 (1H, m, H-3a), 3.45–3.51 (1H, m, H-3b), 3.14 (1H, ddd, J = 16.5, 10.5, 6.0 Hz, H-4a), 2.95 (1H, dt, J = 16.0, 4.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 149.5 (C-1'), 134.5, 131.0, 130.6, 129.3, 129.2, 129.0, 127.1, 126.2, 117.5 (C≡N), 116.2, 95.3, 52.5 (C-1), 44.1 (C-3), 28.4 (C-4); positive ESI-MS m/z: 334 [M−CN]+, 383 [M+Na]+. HR-MS [M+Na]+: Calcd. for C16H13IN2Na+, 383.0021, found 383.0017.
2-(4-Iodophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (12): white powder; yield: 88%; m.p. 145–147 °C; IR (KBr) υmax cm−1: 2221 (w, C≡N), 1140 (s, C−N); UV (MeOH): λmax (lg ε) 258 (4.39) nm; 1H-NMR (CDCl3) δ: 7.66 (2H, d-like, J = 9.0 Hz, H-3', H-5'), 7.31–7.34 (3H, m), 7.26–7.28 (1H, m), 6.86 (2H, d-like, J = 9.0 Hz, H-2', H-6'), 5.49 (1H, s, H-1), 3.73–3.78 (1H, m, H-3a), 3.47–3.52 (1H, m, H-3b), 3.17 (1H, ddd, J = 16.5, 10.5, 6.0 Hz, H-4a), 3.01 (1H, dt, J = 16.0, 4.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 148.0, 138.4 (C-3', C-5'), 134.5, 129.4, 129.2, 129.0, 127.1, 127.0, 119.3, 117.5 (C≡N), 84.2, 52.5 (C-1), 44.1 (C-3), 28.4 (C-4); positive ESI-MS m/z: 334 [M−CN]+, 361 [M+H]+, 383 [M+Na]+. HR-MS [M+Na]+: Calcd. for C16H13IN2Na+, 383.0021, found 383.0014.
2-(2-(Trifluoromethyl)phenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (13): white powder; yield: 90%; m.p. 101–102 °C; IR (KBr) υmax cm−1: 2224 (w, C≡N), 1169 (s, C−N), 1312 (s, C−F); UV (MeOH): λmax (lg ε) 245 (4.05) nm; 1H-NMR (CDCl3) δ: 7.70 (2H, d, J = 8.0 Hz), 7.65 (1H, t, J = 7.5 Hz), 7.39 (1H, t, J = 7.5 Hz), 7.31 (1H, t, J = 7.5 Hz), 7.20–7.26 (3H, m), 5.13 (1H, s, H-1), 3.65–3.71 (1H, m, H-3a), 3.20–3.28 (2H, m, H-3b, H-4a), 2.87–2.91 (1H, m, H-4b); 13C-NMR (CDCl3) δ: 148.3, 134.1, 133.4, 129.8, 129.7, 128.6, 127.3 (q, J = 5.0 Hz), 126.9 (d, J = 7.5 Hz), 126.6, 126.4, 117.9 (C≡N), 56.2 (C-1), 47.2 (C-3), 28.9 (C-4); positive ESI-MS m/z: 276 [M−CN]+, 325 [M+Na]+. HR-MS [M+Na]+: Calcd. for C17H13F3N2Na+, 325.0929, found 325.0933.
2-(3-(Trifluoromethyl)phenyl)-1,2-dihydroisoquinoline-1-carbonitrile (14): white crystal; yield: 93%; m.p. 100–101 °C; IR (KBr) υmax cm−1: 2226 (w, C≡N), 1166 (s, C−N), 1327 (s, C−F); UV (MeOH): λmax (lg ε) 251 (4.64) nm; 1H-NMR (CDCl3) δ: 7.47 (1H, t, J = 8.0 Hz), 7.30–7.35 (3H, m), 7.22–7.27 (4H, m), 5.54 (1H, s, H-1), 3.79–3.83 (1H, m, H-3a), 3.54 (1H, td-like, J = 10.0, 4.0 Hz, H-3b), 3.17 (1H, ddd, J = 16.0, 10.0, 6.0 Hz, H-4a), 3.02 (1H, dt, J = 16.5, 4.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 148.5, 134.4, 132.0 (d, J = 32.0 Hz), 130.2, 129.4, 129.1, 129.0, 119.8, 118.0, 117.4 (C≡N), 113.7, 52.4 (C-1), 44.2 (C-3), 28.4 (C-4); positive ESI-MS m/z: 276 [M−CN]+. HR-MS [M+Na]+: Calcd. for C17H13F3N2Na+, 325.0929, found 325.0939.
2-(4-(Trifluoromethyl)phenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (15): white powder; yield: 98%; m.p. 101–102 °C; IR (KBr) υmax cm−1: 2224 (w, C≡N), 1172 (s,C−N), 1326 (s, C−F); UV (MeOH): λmax (lg ε) 205 (4.40), 259 (4.32) nm; 1H-NMR (CDCl3) δ: 7.61 (2H, d, J = 8.5 Hz, H-3', H-5'), 7.26–7.37 (4H, m), 7.09 (2H, d, J = 8.5 Hz, H-2', H-6'), 5.58 (1H, s, H-1), 3.84–3.88 (1H, m, H-3a), 3.55–3.60 (1H, m, H-3b), 3.17 (1H, ddd, J = 16.0, 10.0, 6.0 Hz, H-4a), 3.06 (1H, dt, J = 16.0, 8.5, 4.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 150.4 (C-1'), 134.6, 129.3, 129.1, 129.1, 127.2, 127.1, 126.9, 126.9, 117.5 (C≡N), 115.4, 51.3 (C-1), 44.0 (C-3), 28.3 (C-4); positive ESI-MS m/z: 276 [M−CN]+, 325 [M+Na]+. HR-MS [M+Na]+: Calcd. for C17H13F3N2Na+, 325.0929, found 325.0935.
2-(3-Nitrophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (16): yellow powder; yield: 90%; m.p. 168–170 °C; IR (KBr) υmax cm−1: 2224 (w, C≡N), 1149 (s, C−N), 1579 (s, NO2); UV (MeOH): λmax (lg ε) 211 (4.13), 247 (4.31) nm; 1H-NMR (CDCl3) δ: 7.89 (1H, s, H-2'), 7.83 (1H, d, J = 8.0 Hz), 7.52 (1H, t, J = 8.0 Hz), 7.32–7.36 (4H, m), 7.26–7.29 (1H, m), 5.60 (1H, s, H-1), 3.85–3.89 (1H, m, H-3a), 3.57–3.62 (1H, m, H-3b), 3.19 (1H, ddd, J = 16.0, 10.0, 6.0 Hz, H-4a), 3.08 (1H, dt, J = 16.0, 4.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 148.9 (C-1'), 134.4 (C-3'), 130.4, 129.3, 129.2, 128.8, 127.3, 127.1, 121.7, 117.2 (C≡N), 115.7, 111.0, 51.7 (C-1), 44.1 (C-3), 28.3 (C-4); positive ESI-MS m/z: 253 [M−CN]+, 302 [M+Na]+. HR-MS [M+Na]+: Calcd. for C16H13N3NaO2+, 302.0905, found 302.0900.
2-(4-Nitrophenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (17): yellow powder; yield: 93%; m.p. 102–104 °C; IR (KBr) υmax cm−1: 2361 (w, C≡N), 1162 (s, C−N), 1597 (s, NO2); UV (MeOH): λmax (lg ε) 368 (4.00) nm; 1H-NMR (CDCl3) δ: 8.16–8.18 (2H, d, J = 9.0 Hz, H-2', H-6'), 7.25–7.33 (4H, m), 6.94–6.98 (2H, d, J = 8.5 Hz, H-3', H-5'), 5.79 (1H, s, H-1), 3.77–3.81 (1H, m, H-3a), 3.50–3.56 (1H, m, H-3b), 3.38–3.44 (1H, m, H-4a), 2.97 (1H, dt, J = 15.0, 5.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 153.6 (C-1'), 138.9 (C-4'), 135.7, 133.6, 129.0, 128.0, 126.5, 126.1, 126.0, 113.4 (C≡N), 112.3, 61.4 (C-1), 43.7 (C-3), 27.8 (C-4); positive ESI-MS m/z: 253 [M−CN]+. HR-MS [M+Na]+: Calcd. for C16H13N3NaO2+, 302.0905, found 302.0901.
2-(2-Methylphenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (18): yellow crystal; yield: 82%; m.p. 136–138 °C; IR (KBr) υmax cm−1: 2220 (w, C≡N), 1141 (s, C−N); UV (MeOH): λmax (lg ε) 237 (3.88) nm; 1H-NMR (CDCl3) δ: 7.22–7.31 (7H, m), 7.14–7.11 (1H, m), 5.06 (1H, s, H-1), 3.61 (1H, td, J = 11.4, 3.8 Hz, H-3a), 3.35–3.32 (1H, m, H-3b), 3.18 (1H, ddd, J = 16.8, 11.4, 6.2 Hz, H-4a), 2.92 (1H, d-like, J = 16.8 Hz, H-4b), 2.29 (3H, s); 13C-NMR (CDCl3) δ: 148.0 (C-1'), 134.5, 133.4, 131.2, 130.1, 129.7, 128.6, 127.2, 127.0, 126.6, 125.4, 122.0, 117.7 (C≡N), 55.6 (C-1), 44.9 (C-3), 28.7 (C-4); positive ESI-MS m/z: 222 [M−CN]+. HR-MS [M+Na]+: Calcd. for C17H16N2Na+, 271.1211, found 271.1207.
2-(3-Methylphenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (19): yellow oil; yield: 75%; IR (KBr) υmax cm−1: 2224 (w, C≡N), 1030 (vs, C−N); UV (MeOH): λmax (lg ε) 247 (4.12) nm; 1H-NMR (CDCl3) δ: 7.28–7.20 (5H, m), 6.88–6.86 (2H, m), 6.82 (1H, d, J = 7.5 Hz, H-4'), 5.49 (1H, s, H-1), 3.74 (1H, ddd, J = 12.4, 6.0, 2.9 Hz, H-3a), 3.44 (1H, ddd, J = 12.4, 10.9, 4.1 Hz, H-3b), 3.12 (1H, ddd, J = 16.4, 10.9, 6.0 Hz, H-4a), 2.92 (1H, dt, J = 16.4, 3.4 Hz, H-4b), 2.35 (3H, s); 13C-NMR (CDCl3) δ: 148.5 (C-1'), 139.4, 134.7, 129.7, 129.5, 129.4, 128.8, 127.1, 126.9, 122.8, 118.4, 117.9 (C≡N), 114.7, 53.3 (C-1), 44.2 (C-3), 28.6 (C-4); positive ESI-MS m/z: 222[M−CN]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literatural data [17].
2-(4-Methylphenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (20): yellow solid; yield: 64%; m.p. 114–116 °C; IR (KBr) υmax cm−1: 2222 (w, C≡N), 1137 (s, C−N); UV (MeOH): λmax (lg ε) 245 (4.11) nm; 1H-NMR (CDCl3) δ: 7.30–7.21 (4H, m), 7.16 (2H, d, J = 8.2 Hz, H-3', 5'), 7.00 (2H, d, J = 8.4 Hz, H-2', 6'), 5.45 (1H, s, H-1), 3.70 (1H, ddd, J = 12.4, 6.1, 2.3 Hz, H-3a), 3.44 (1H, ddd, J = 12.4, 11.2, 4.0 Hz, H-3b), 3.15 (1H, ddd, J = 16.7, 11.2, 6.1 Hz, H-4a), 2.94 (1H, dt, J = 16.7, 3.2 Hz, H-4b), 2.31 (3H, s); 13C-NMR (CDCl3) δ: 146.3 (C-1'), 134.6, 131.8, 130.1 (C-3', 5'), 129.7, 129.4, 128.7, 127.1, 126.8, 118.3 (C-2', 6'), 117.7 (C≡N), 114.8, 54.1 (C-1), 44.4 (C-3), 28.6 (C-4); positive ESI-MS m/z: 222 [M−CN]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-(2-Methoxyphenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (21): white solid; yield: 67%; m.p. 150–152°C; IR (KBr) υmax cm−1: 2223 (w, C≡N), 1144 (s, C−N), 1243 (vs, C-O-C); UV (MeOH): λmax (lg ε) 241 (3.84), 278 (3.48) nm; 1H-NMR (CDCl3) δ: 7.12–7.30 (6H, m), 7.02 (1H, td, J = 8.0, 1.0 Hz,), 6.92 (1H, d, J = 8.0 Hz), 5.74 (1H, s, H-1), 3.85 (3H, s), 3.52 (2H, dd, J = 8.0 Hz, H-3), 3.20–3.26 (1H, m, H-4a), 2.92 (1H, dt, J = 16.5, 2.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 152.5 (C-2'), 137.8 (C-1'), 134.1, 129.9, 129.6, 128.5, 127.2, 126.6, 125.1, 121.5, 121.0, 117.8 (C≡N), 111.5, 55.6 (C-1), 53.2, 44.8 (C-3), 28.8 (C-4); positive ESI-MS m/z: 238 [M−CN]+, 265 [M+H]+, 287 [M+Na]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-(3-Methoxyphenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (22): yellow oil; yield: 73%; IR (KBr) υmax cm−1: 2221(w, C≡N), 1165 (s, C−N), 1212 (vs, C-O-C)); UV (MeOH): λmax (lg ε) 245 (4.09) nm; 1H-NMR (CDCl3) δ: 7.21–7.31 (5H, m), 6.54–6.68 (3H, m), 5.51 (1H, s, H-1), 3.81 (3H, s), 3.74–3.77 (1H, m, H-3a), 3.43–3.49 (1H, m, H-3b), 3.10–3.16 (1H, m, H-4a), 2.95 (1H, d, J = 16.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 160.8 (C-3'), 149.7 (C-1'), 134.7, 130.4, 129.6, 129.4, 128.8, 127.1, 126.9, 117.8 (C≡N), 109.9, 106.6, 104.0, 55.4(C-1), 53.0, 44.2 (C-3), 28.5 (C-4); positive ESI-MS m/z: 238 [M−CN]+, 265 [M+H]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-(4-Methoxyphenyl)-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (23): yellow oil; yield: 78%; IR (KBr) υmax cm−1: 2222 (w, C≡N), 1136 (s, C−N), 1248 (vs, C-O-C); UV (MeOH): λmax (lg ε) 242 (4.05) nm; 1H-NMR (CDCl3) δ: 7.21–7.30 (4H, m), 7.08 (2H, d, J = 9.0 Hz, H-3', H-5'), 6.91 (2H, d, J = 9.0 Hz, H-2', H-6'), 5.36 (1H, s, H-1), 3.79 (3H, s), 3.55–3.59 (1H, m, H-3a), 3.42 (1H, td, J = 11.5, 3.5 Hz, H-3b), 3.15 (1H, ddd, J = 16.5, 11.5, 6.5 Hz, H-4a), 2.92 (1H, d, J = 16.0 Hz, H-4b); 13C-NMR (CDCl3) δ: 155.7 (C-4'), 142.6 (C-1'), 134.4, 129.7, 129.5, 129.0, 127.1, 126.7, 121.0, 117.7 (C≡N), 114.8, 55.6 (C-1), 44.9 (C-3), 28.7 (C-4); positive ESI-MS m/z: 238 [M−CN]+, 265 [M+H]+, 287 [M+Na]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].
2-Phenyl-1,2,3,4-tetrahydroisoquinoline-1-carbonitrile (24): white solid; yield: 73%; m.p. 89–90 °C; IR (KBr) υmax cm−1: 2223 (s, C≡N), 1222, 1206 (C−N); UV (MeOH): λmax (lg ε) 245 (4.04) nm; 1H-NMR (CDCl3) δ: 7.39–7.43 (2H, m), 7.31–7.37 (3H, m), 7.27–7.30 (1H, m), 7.14 (1H, s), 7.13 (1H,s), 7.06 (1H, t, J = 7.5 Hz), 5.56 (1H, s, H-1), 3.82 (1H, ddd, J = 12.5, 6.0, 3.0 Hz, H-3a), 3.53 (1H, ddd, J = 12.0, 10.5, 4.0 Hz, H-3b), 3.19 (1H, ddd, J = 16.5, 10.5, 6.0 Hz, H-4a), 3.01 (1H, dt, J = 16.0, 3.5 Hz, H-4b); 13C-NMR (CDCl3) δ: 148.4 (C-1'), 134.7, 129.7, 129.4, 128.8, 127.1, 126.9, 121.9, 117.8 (C≡N), 117.6, 53.2 (C-1), 44.2 (C-3), 28.6 (C-4); positive ESI-MS m/z: 208 [M−CN]+. The IR, 1H-NMR and 13C-NMR data are agreement with the literature data [17].

3.3. Acaricidal Activity Assay

The acaricidal activity of compounds 124 was assayed against Psoroptes cuniculi according to our previously reported method [10]. Briefly, adult mites of both sexes were isolated from naturally infected rabbits under a stereoscopic microscope, and placed in 24-well flat-bottomed cell culture plates (20 adult mites/well). The solution of the tested compound with 0.4 mg/mL was prepared in the mixed solution of dimethyl sulfoxide (DMSO), Tween-80 and normal saline (1:1:8, v/v/v). Half a milliliter of the solution was directly added to each well. Three replicates were made for each compound. The same solution without the tested compound was used as untreated control. The solution of ivermectin, a standard acaricide, was used as positive drug control. All plates were placed in separate humidity chambers in saturated humidity conditions at 22 °C. After 24 h, each well was observed under a stereomicroscope and all the motionless mites were stimulated with a needle. Lack of reactions and persistent immobility indicated their death. Mortality was calculated as the following formula and expressed as means ± S.D.:
Molecules 19 08051 i001

3.4. Acaricidal Toxicity Assay

According to our previously reported method [9], compound 1 with the strongest activity was selected to determine its acaricidal toxicity against P. cuniculi. A 2.0 mg/mL stock solution of the tested compound was prepared in a mixture of dimethyl sulfoxide (DMSO), Tween-80 and normal saline (1:1:8, v/v/v), and then diluted with the same mixed solvent to obtain a series of concentrations of 1.8, 1.6, 1.4, 1.2, 1, 0.5, 0.25, 0.125, 0.0625, 0.0313, 0.0157 and 0.0078 mg/mL. The acaricidal activity for each concentration was tested according to the same procedure described above. Each test was performed in triplicate. The mortality of mites for each test was calculated and then corrected by applying Abbott’s formula:
Molecules 19 08051 i002
Probit value of the corrected mortality for each test concentration and the corresponding log[concentration (mg/L)] were used to establish toxicity regression equation with the linear least-square fitting method. The LC50 value and its confidence interval at 95% probability were calculated from the toxicity regression equation.
Test solutions of 1 or ivermectin (3 mg/mL) in the same mixed solvent was prepared and used to determine the LT50 value. The determination of acaricidal activity of each solution was carried out according to the method described above. The mites in each well were observed under a stereomicroscope every 0.5 h or 1.0 h and the percentage mortality and corrected percentage mortality for each well in each set time was calculated. The tests were performed in triplicate. The corrected percentage mortality at each set time was expressed as means ± S.D. Toxicity regression equation for time effect was established between the probit value of the corrected percentage mortality for each post-treatment time and the corresponding log[post-treatment time (h)], and used to calculate LT50.

4. Conclusions

In conclusion, we have reported the synthesis of a series of the target compounds and their in vitro acaricidal activity against P. cuniculi, and also discussed their SAR. All compounds 124 were found to possess different degrees of activity at 0.4 mg/mL. Among them, 1 displayed the highest activity with an LC50 value of 0.2421 μg/mL and an LT50 value of 7.79 h, and an average mortality rate of 66.7%, superior to the standard drug ivermectin. The SAR showed that 2'-F, 3'-F, 2'-Cl, 2'-Br and 2'-CF3 substituents on the N-aromatic ring remarkably enhanced the activity. Thus, the target compounds can be considered a novel class of promising candidates or lead compounds for the development of new tetrahydroisoquinoline acaricidal agents.

Acknowledgments

This project was supported by the National Natural Science Foundation of China (NSFC: No. 31172365, 31101469), New Century Excellent Talents by Ministry of Education of China (NCET-12-0475), the Fund of Youth Science and Technology Stars by Shaanxi Province (2012KJXX-16), and the Fundamental Research Funds for the Central Universities (QN2011035).

Author Contributions

Yang R. and Qiao R. designed and synthesized the title compounds; Zhang B.Y., Zheng Z.L. and Fang M. carried out the bioactivity assay; Zhou L. and Geng H.L. were responsible for the identification of all compounds and compiled the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bates, P.G. Inter- and intra-specific variation within the genus Psoroptes (Acari: Psoroptidae). Vet. Parasitol. 1999, 83, 201–217. [Google Scholar] [CrossRef]
  2. Flatt, R.E.; Wiemers, J. A survey of fur mites in domestic rabbits. Lab. Anim. Sci. 1976, 26, 758–761. [Google Scholar]
  3. Yeatts, J.W. Rabbits mite infestation. Vet. Rec. 1994, 134, 359–360. [Google Scholar]
  4. Synge, B.A.; Bates, P.G.; Clark, A.M.; Stephen, F.B. Apparent resistance of P. ovis to flumethrin. Vet. Rec. 1995, 137, 51. [Google Scholar]
  5. Clark, A.M.; Stephen, F.B.; Cawley, G.D.; Bellworthy, S.J.; Groves, B.A. Resistance of the sheep scab mite Psoroptes ovis to propetamphos. Vet. Rec. 1996, 139, 451. [Google Scholar]
  6. Currie, B.J.; Harumal, P.; McKinnon, M.; Walton, S.F. First documentation of in vivo and in vitro ivermectin resistance in Sarcoptes scabiei. Clin. Infect. Dis. 2004, 39, e8–e12. [Google Scholar] [CrossRef]
  7. Halley, B.A.; VandenHeuvel, W.J.A.; Wislocki, P.G. Enviromental effects of the usage of avemectins in livestock. Vet. Parasitol. 1993, 48, 109–125. [Google Scholar] [CrossRef]
  8. O’Brien, D.J. Treatment of psoroptic mange with reference to epidemiology and history. Vet. Parasitol. 1999, 83, 177–185. [Google Scholar] [CrossRef]
  9. Miao, F.; Yang, X.J.; Ma, Y.N.; Zheng, F.; Song, X.P.; Zhou, L. Structural modification of sanguinarine and chelerythrine and their in vitro acaricidal activity against Psoroptes cuniculi. Chem. Pharm. Bull. 2012, 60, 1508–1513. [Google Scholar]
  10. Ma, Y.N.; Yang, X.J.; Pan, L.; Hou, Z.; Geng, H.L.; Song, X.P.; Zhou, L.; Miao, F. Synthesis of 2-aryl-3,4-dihydroisoquinolin-2-ium bromides and their in vitro acaricidal activity against Psoroptes cuniculi. Chem. Pharm. Bull. 2013, 61, 204–211. [Google Scholar] [CrossRef]
  11. Alexis, D.C.; Parenty, A.D.C.; Smith, L.V.; Guthrie, K.M.; Long, D.-L.; Plumb, J.; Brown, R.; Cronin, L. Highly stable phenanthridinium frameworks as a new class of tunable DNA binding agents with cytotoxic properties. J. Med. Chem. 2005, 48, 4504–4506. [Google Scholar] [CrossRef]
  12. Nakanishi, T.; Masuda, A.; Suwa, M.; Akiyama, Y.; Hoshino-Abe, N.; Suzuki, M. Synthesis of derivatives of NK109, 7-OH benzo[c]phenanthridine alkaloid, and evaluation of their cytotoxicities and reduction-resistant properties. Bioorg. Med. Chem. Lett. 2000, 10, 2321–2323. [Google Scholar] [CrossRef]
  13. Eldin, S.; Jencks, W.P. Lifetimes of iminium ions in aqueous solution. J. Am. Chem. Soc. 1995, 117, 4851–4857. [Google Scholar] [CrossRef]
  14. Parenty, A.D.C.; Smith, L.V.; Pickering, A.L.; Long, D.L.; Cronin, L. General one-pot, three-step methodology leading to an extended class of N-heterocyclic cations: Spontaneous nucleophilic addition, cyclization, and hydride loss. J. Org. Chem. 2004, 69, 5934–5946. [Google Scholar] [CrossRef]
  15. Shimizu, M.; Itou, H.; Miura, M. A new synthetic method for á-alkoxycarbonyl iminium salt and its reaction with nucleophiles. J. Am. Chem. Soc. 2005, 127, 3296–3297. [Google Scholar] [CrossRef]
  16. Yoshida, J.; Suga, S.; Suzuki, S.; Kinomura, N.; Yamamoto, A.; Fujiwara, K. Direct oxidative carbon-carbon formation using the “cation pool” method. 1. Generation of iminium cation pools and their reactions with carbon nucleophiles. J. Am. Chem. Soc. 1999, 121, 9546–9549. [Google Scholar] [CrossRef]
  17. Zhang, G.; Ma, Y.X.; Cheng, G.B.; Liu, D.B.; Wang, R. A unique combined source of “CN” from 1,2-dichloroethane and TMSN3 in the copper-catalyzed cyanation of a C(sp3)−H bond adjacent to a nitrogen atom. Org. Lett. 2014, 16, 656–659. [Google Scholar] [CrossRef]
  18. Dostál, J.; Slavík, J. Some aspects of the chemistry of quaternary benzo[c]phenanthridine alkaloids. Stud. Nat. Prod. Chem. 2002, 27, 155–184. [Google Scholar] [CrossRef]
  19. Walterová, D.; Ulrichová, J.; Válka, I.; Vicar, J.; Vavrecková, C.; Táborská, E.; Harjrader, R.J.; Meyer, D.L.; Cerná, H.; Simánek, V. Benzo[c]phenanthridine alkaloids sanguinarine and chelerythrine: Biological activities and dental care applications. Acta Univ. Palacki. Olomuc. Fac. Med. 1995, 139, 7–16. [Google Scholar]
  20. Seèkáøová, P.; Marek, R.; Dostál, J.; Dommisse, R.; Esmans, E.L. Structural studies of benzophenanthridine alkaloid free bases by NMR spectroscopy. Magn. Reson. Chem. 2002, 40, 147–152. [Google Scholar] [CrossRef]
  • Sample Availability: Samples of the compounds 124 are available from the authors.

Share and Cite

MDPI and ACS Style

Yang, R.; Ruan, Q.; Zhang, B.-Y.; Zheng, Z.-L.; Miao, F.; Zhou, L.; Geng, H.-L. A Class of Promising Acaricidal Tetrahydroisoquinoline Derivatives: Synthesis, Biological Evaluation and Structure-Activity Relationships. Molecules 2014, 19, 8051-8066. https://doi.org/10.3390/molecules19068051

AMA Style

Yang R, Ruan Q, Zhang B-Y, Zheng Z-L, Miao F, Zhou L, Geng H-L. A Class of Promising Acaricidal Tetrahydroisoquinoline Derivatives: Synthesis, Biological Evaluation and Structure-Activity Relationships. Molecules. 2014; 19(6):8051-8066. https://doi.org/10.3390/molecules19068051

Chicago/Turabian Style

Yang, Rui, Qiao Ruan, Bing-Yu Zhang, Zuo-Lue Zheng, Fang Miao, Le Zhou, and Hui-Ling Geng. 2014. "A Class of Promising Acaricidal Tetrahydroisoquinoline Derivatives: Synthesis, Biological Evaluation and Structure-Activity Relationships" Molecules 19, no. 6: 8051-8066. https://doi.org/10.3390/molecules19068051

Article Metrics

Back to TopTop