Next Article in Journal
Changes in Arsenic and Copper Bioavailability and Oxytetracycline Degradation during the Composting Process
Next Article in Special Issue
The Masked Polar Group Incorporation (MPGI) Strategy in Drug Design: Effects of Nitrogen Substitutions on Combretastatin and Isocombretastatin Tubulin Inhibitors
Previous Article in Journal
Experimental Cannabinoid 2 Receptor Activation by Phyto-Derived and Synthetic Cannabinoid Ligands in LPS-Induced Interstitial Cystitis in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design and Synthesis of New 6-Nitro and 6-Amino-3,3a,4,5-Tetrahydro-2H-Benzo[g]indazole Derivatives: Antiproliferative and Antibacterial Activity

by
Viviana Cuartas
1,2,
María del Pilar Crespo
3,
Eva-María Priego
4,
Leentje Persoons
5,
Dirk Daelemans
5,
María-José Camarasa
4,
Braulio Insuasty
1,2,* and
María-Jesús Pérez-Pérez
4,*
1
Grupo de Investigación de Compuestos Heterocíclicos, Departamento de Química, Universidad del Valle, A. A. Cali 25360, Colombia
2
Centre for Bioinformatics and Photonics-CIBioFI, Calle 13 No. 100-00, Edificio E20, No. 1069, Cali 760032, Colombia
3
Grupo de Biotecnología e Infecciones Bacterianas, Departamento de Microbiología, Universidad del Valle, Cali 760043, Colombia
4
Instituto de Química Médica (IQM, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
5
KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
*
Authors to whom correspondence should be addressed.
Molecules 2019, 24(23), 4236; https://doi.org/10.3390/molecules24234236
Submission received: 30 October 2019 / Revised: 15 November 2019 / Accepted: 19 November 2019 / Published: 21 November 2019
(This article belongs to the Special Issue Tubulin Inhibitors)

Abstract

:
New substituted benzo[g]indazoles functionalized with a 6-nitro and 6-amino groups have been synthesized by the reaction of benzylidene tetralones with hydrazine in acetic acid. The resulting conformationally-constrained compounds were evaluated for their antiproliferative activity against selected cancer cell lines. The nitro-based indazoles 11a, 11b, 12a and 12b have shown IC50 values between 5–15 μM against the lung carcinoma cell line NCI-H460. Moreover, the nitro compounds were tested for antibacterial activity where compounds 12a and 13b have shown MIC values of 250 and 62.5 μg/mL against N. gonorrhoeae with no hemolytic activity in human red blood cells (RBC).

Graphical Abstract

1. Introduction

Indazoles are benzo-fused pyrazoles for which a broad range of biological properties have been described [1,2]. Fused tricyclic pyrazole derivatives have been studied as necroptosis inhibitors [3,4], high affinity ligands for the human dopamine D4 receptor [5], phosphodiesterase 4 (PDE4) inhibitors [6], cannabinoid-2 receptor ligands [7], inhibitors of β-hematin formation conferring in vitro antimalarial activity [8] or inhibition of isocitrate dehydrogenase 1 (IDH1) [9]. Moreover, the tricyclic pyrazole core is present in compounds with antimicrobial [10,11,12] and antiproliferative activity [13,14,15]. The most widely used synthetic approach towards these compounds involves the reaction of α,β-unsaturated ketones and hydrazines [6,8,10,11,12,14]. Very recently, a Lewis acid promoted reaction of cycloalkanones with hydrazones has also been described to synthesize 2,3-diaryl-4,5,6,7-tetrahydro-1H-indazoles [16]. It should be highlighted that these fused tricyclic pyrazole derivatives are characterized by their conformational restriction [15].
Colchicine-site inhibitors based on combretastatin A-4 (1, CA-4, Figure 1A) have been extensively studied as antimitotic and vascular disrupting agents [17,18,19]. Among the different approaches followed to mimic the cis (active) configuration of CA-4, the design and synthesis of conformationally restricted ligands have led to very potent inhibitors. Among recently reported examples, the dihydronaphthalene and benzosuberene derivatives (2 and 3, respectively, Figure 1A) have shown antiproliferative activity at the sub nM range [20]. Very recently, Jiang, J. et al. [21] described a novel 1-phenyl-dihydrobenzoindazole (4, Figure 1A) with a locked conformation, that inhibited tubulin polymerization with an IC50 of 1.6 μM and showed antitumor properties against a human colon cancer cell line with an IC50 value of 1 nM. In both series the presence of an amino group on ring B is meant to mimic the phenolic OH of CA-4 and this amino group has been used for the synthesis of prodrugs thereof [20,21]. In addition, in all cases a trimethoxyphenyl is present as ring A (Figure 1).
We have recently reported on tetrahydronaphtalene derivatives as conformational restricted mimetics of chalcone derivatives that also bind the colchicine binding site in tubulin [22]. As in the previous examples, an amino group was incorporated at position 5 on ring B and this was shown to be crucial for the antiproliferative activity and the tubulin binding capacity. Thus, compound 5 was found to inhibit CEM (human lymphoblastic leukemia) proliferation at the sub µM range.
Based on the general interest of tetrahydroindazole derivatives, we have here designed and synthesized a new series of tricyclic pyrazoline derivatives incorporating an amino group at position 6 on ring B that were obtained through reduction of their nitro precursors (Figure 1B). Several 4-methoxy aryl rings have been incorporated as ring A. In particular, those compounds with a 3,4,5-trimethoxyphenyl as ring A and an amino group at position 6 on ring B could mimic the conformation of our colchicine-binding site inhibitor 5, as will be later discussed. The synthesized compounds have been tested for their antiproliferative activity against a wide panel of tumor cells. As a part of our continuing effort to find molecules with antibacterial activity [23,24,25,26], the nitro derivatives were also screened against wild type and multidrug-resistant bacteria, particularly those frequently found in relevant human infections.

2. Results and Discussion

2.1. Chemistry

The synthesis of the targeted compounds showed in Figure 1B required the 6-methoxy-5-nitro-1-tetralone (6) as a starting material that was prepared as previously described [20,27]. Reaction of 6 by Claisen-Schmidt condensation with aromatic aldehydes (Scheme 1) in the presence of NaOH in ethanol provided the corresponding chalcones 710 in excellent yields (82, 86, 82 and 77% yield, respectively).
The chalcones 79 were then treated with hydrazine hydrate in the presence of acetic acid to give a mixture of diastereoisomers 11a13a and 11b13b in a 1:1 ratio that could be separated by chromatography, and with global yields of 59, 74 and 71%, respectively (Scheme 2). The structural and stereochemical assignment of each pair of enantiomers was performed based on NMR experiments. As an example, the 2D NOESY spectrum for compound 11b showed a NOE signal between the protons H-3 and H-3a, and therefore 11b was assigned as the (3R,3aR)-rel stereoisomer. In addition, the signal corresponding to H-3 in 11b has a coupling constant (J3,3a = 11.0 Hz) larger than the one of the same signal in the diastereomer 11a (J3,3a = 9.7 Hz). When the 13C NMR spectra of 11a and 11b were compared, the signal corresponding to C-3a appeared at 53.8 and 46.9 ppm, respectively. Thus, there is a significant upfield shift for this signal in 11b, suggesting the cis configuration ((3R,3aR)-rel), in agreement with those reported in the literature [8,10,12,28]. Interestingly, in the whole series, one of the signals corresponding to H-4 for the (3R,3aR)-rel isomers showed a chemical shift around 0.7–0.8 ppm, probably due to the shielding effect of the aromatic ring A, while this signal for the (3R,3aS)-rel isomers is around 1.90 ppm. We consider that this difference can also be employed as useful and convenient criteria to assign both isomers. Similarly, reaction of chalcone 10 with hydrazine hydrate in the presence of acetic acid gave a mixture of the two trimethoxy derivatives 14a and 14b (Scheme 2), in a global yield of 66%, but whose separation by chromatography was very laborious. Thus, the mixture of 14a and 14b was used for the next reaction.
Finally, the nitro derivatives 1114a,b were reduced to their amino analogs by treatment with Fe in an EtOH:H2O mixture at reflux in the presence of HCl (Scheme 2), following the procedure previously used for the reduction of the 5-nitrotetrahydronaphthalenes [22], providing compounds 15a18a and 15b18b in moderate to good yields (25–75%). These compounds 15a18a and 15a18b showed similar spectroscopic data and therefore, we discuss here compound 15a as the representative of this series. In the 1H-NMR spectrum, the signal corresponding to H-3a appears as a double-double-doublet at 3.05 ppm with coupling constants 3J = 13.8, 9.2, 4.8 Hz; the signals for H-4 appear as two multiplets at 1.83 ppm integrating for one proton and between 2.15 and 2.17 ppm for four protons corresponding to H-4 and the methyl protons of acetyl group. A broad singlet for amino protons was observed at 4.61 ppm. The 13C-NMR spectrum showed the expected nineteen signals for the carbons of compound 15a.
As mentioned in the introduction, these compounds are characterized by a high degree of conformational restriction, thus they could represent an alternative scaffold for colchicine site binding agents. Thus, 3D structures of compounds 18a and 18b were constructed and their geometries were optimized with MOPAC2012 (AM1 method) [29]. This was followed by a manual superposition of the geometry-optimized structures with the conformation of the tetrahydronaphtalene derivative 5 for which we have recently reported its tubulin binding capacity and antiproliferative activity [22]. As shown in Figure 2, the degree of overlapping is quite relevant, the major differences being the relative orientation of ring A and the presence of the additional acetylated pyrazoline ring.
Interestingly, a comparison of the 3D structures of 18a (3R,3aS) and 18b (3R,3aR) also helps to explain the differences observed in the chemical shifts in the 1H-NMR spectrum of one of the protons H-4. As shown in Figure 3, for 18b one of the H-4 protons is facing the phenyl ring A. Thus, this shielding effect explains the differences in chemical shifts for this proton between the b-isomers (0.7–0.8 ppm) and the a-isomers (around 1.90 ppm) in the whole series.

2.2. Biological Evaluation

2.2.1. Antiproliferative Activity

All benzo[g]indazole derivatives 1118a,b were tested for their antiproliferative activity against a wide panel of tumor cells. As shown in Table 1, most of the 3,3a,4,5-tetrahydro-2H-benzo[g]indazole derivatives showed modest antiproliferative activity against the cell lines tested. In particular, the trimethoxy derivatives 18a and 18b showed IC50 values around 50 µM. On the other hand, the 6-nitro derivatives 11a, 11b and 12a, 12b, showed IC50 values in the low µM range against a lung carcinoma cell line NCI-H460. Some additional studies were performed for compounds 11a, 11b, and 12a, 12b. At 10 µM, the compounds did not affect tubulin as measured by immunostaining in HeLa cells, suggesting that tubulin is not the target for the antiproliferative activity. They were also screened for DNA intercalation or DNA damage, using standardized procedures. These protocols are included as Supplementary Materials. Also, in this case, no significant effect was observed.

2.2.2. Antibacterial Activity

Compounds 710 and 1114a,b were investigated for their antibacterial activity against wild type and multidrug-resistant bacteria including methicillin-susceptible S. aureus ATCC 25923 (MSSA), methicillin-resistant S. aureus ATCC 43300 (MRSA), vancomycin-intermediate S. aureus (VISA), E. coli ATCC 25922, P. aeruginosa ATCC 27853, K. pneumoniae ATCC 700603 (Extended spectrum beta-lactamase positive, ESBL+), K. pneumoniae BAA1705 (carbapenemase-positive) and N. gonorrhoeae ATCC 49226. The minimum inhibitory concentration (MIC) was determined by broth microdilution in those compounds with reproducible antimicrobial effect.
Table 2 shows that N. gonorrhoeae growth was inhibited by 12a and 13b. Compound 13b was the most active of the series with a MIC value of 62.5 μg/mL, whereas 12a induced moderate growth inhibition (MIC = 250 μg/mL). Comparison of the antibacterial activity of the isomers 13a (3R,3aS)-rel- (MIC ≥ 500 μg/mL) and 13b (3R,3aR)-rel- (MIC = 62.5 μg/mL) against N. gonorrhoeae shows that the configuration of the molecule has an important effect on the activity. On the other hand, it is worth to take into account the recent emergence of resistant clinical isolates of N. gonorrhoeae to a variety of antimicrobial agents, for which the need to find new antigonococcal agents is of paramount importance [30,31,32]. Compounds 12a and 13b were further tested for hemolytic activity or toxicity against RBC. None of the active compounds induced hemolysis of the human RBC.

3. Materials and Methods

3.1. General Information

Reagents and solvents used were purchased from Sigma-Aldrich (St. Louis, MO, USA). Reactions were monitored by TLC on silica gel 60 F254 aluminum plates (Merck, Dramstand, Germany). Spots were detected under UV light (254 nm) and/or charring with ninhydrin or phosphomolybdic acid. Melting points were measured on a M170 apparatus (Mettler Toledo, Columbus, Ohio, USA) and are uncorrected. The elemental analyses were performed using a CHN-O-RAPID instrument (Heraeus, Hanau, Germany). Analyses indicated by the symbols of the elements or functions were within ±0.4% of the theoretical values. 1H and 13C-NMR spectra were run on an INOVA-300 instrument (Varian, now Agilent, Santa Clara, CA, USA) operating at 300 (1H) and 75 MHz (13C) and a Varian INOVA-400 operating at 400 (1H) and 100 MHz (13C), respectively. Chemical shift (δ) values are reported in parts per million (ppm). Separations on silica gel were performed by preparative centrifugal circular thin-layer chromatography (CCTLC) on a ChromatotronR (Kiesegel 60 PF254 gipshaltig, Merck), with a layer thickness of 1 and 2 mm and flow rate of 4 or 8 mL/min, respectively. Flash chromatography was performed on silica gel as stationary phase. HPLC analysis was performed on an Agilent 1120 compact LC system (Santa Clara, CA, USA), equipped with an ACE 5 C18-300 column (15 cm × 4.6 mm), UV detection, and flow rate 1 mL/min, using as mobile phase A H2O (containing 0.1% TFA) and as mobile phase B acetonitrile. Retention times (TR) are reported in minutes. For all compounds, HPLC purity was determined to be greater than 95%. Compounds were also analyzed by HPLC/MS with an e2695 LC (Waters, Milford, Massachusetts, USA) coupled to a Waters 2996 Photodiode Array Detector and a Waters Micromass ZQ. The column used is a Waters SunFire C18 2.1 × 50 mm, 3.5 µm, and the mobile phases were A: acetonitrile and B: H2O, together with a constant 5% of C (H2O with 2% formic acid) to assure a 0.1% of formic acid along the run.

3.2. Chemistry

3.2.1. Preparation of 6-Methoxy-5-nitro-1-tetralone (6)

To a solution of 6-methoxy-1-tetralone (600 mg, 3.40 mmol) in acetic anhydride (3.0 mL) in an ice bath, a mixture of 65% HNO3 (330 µL) in acetic acid (210 µL) was added dropwise over 2 h, and then the mixture was stirred for additional 2 h at 0 °C. The resulting solid was treated with H2O (20 mL) and extracted with EtOAc (3 × 30 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure. The residue was purified by silica gel chromatography (hexane:EtOAc 8:2) to provide 252 mg (33%) of 6-methoxy-5-nitro-1-tetralone, and 233 mg (31%) of 6-methoxy-7-nitro-1-tetralone as light yellow solids, respectively. Data for 6-methoxy-5-nitro-1-tetralone: 1H-NMR (DMSO-d6, 400 MHz) δ: 2.05 (p, J = 6.5 Hz, 2H), 2.60 (t, J = 6.5 Hz, 2H), 2.78 (t, J = 6.5 Hz, 2H), 3.97 (s, 3H, OCH3), 7.36 (d, J = 8.9 Hz, 1H, Ar), 8.08 (d, J = 8.9 Hz, 1H, Ar). Mp 166–167 °C. 1H-NMR data are similar to those previously described [20,27].

3.2.2. General Procedure for the Synthesis of Chalcones 710 Derived from 6-Methoxy-5-Nitro-1-Tetralone (General Procedure A)

A mixture of 6-methoxy-5-nitro-1-tetralone (6) (1.0 eq), the selected aldehyde (1.2 eq) and a pellet of NaOH (185 mg) in ethanol was stirred at room temperature for 7 h. The solid thus formed was filtered and washed with ethanol and water to provide the chalcones 7 and 8 as pure yellow solids. For 9 and 10 purifications by flash column chromatography were required and performed as specified.
(E)-6-Methoxy-2-(4-methoxybenzylidene)-5-nitro-3,4-dihydronaphthalen-1(2H)-one (7), following the general procedure A, 6-methoxy-5-nitro-1-tetralone (6) (100 mg, 0.45 mmol) reacted with p-anisaldehyde (79 mg, 0.58 mmol) in ethanol (3.0 mL). The precipitate was collected by filtration and washed with ethanol and water to give 125 mg (81% yield) of 7, which did not require further purification. Mp 171–173 °C. MS (ES, positive mode): m/z 340 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 2.79 (t, J = 6.5 Hz, 2H, H-4), 3.11 (t, J = 6.5 Hz, 2H, H-3), 3.81 (s, 3H, OCH3), 3.99 (s, 3H, OCH3), 7.03 (d, J = 8.7 Hz, 2H, Hm), 7.40 (d, J = 8.9 Hz, 1H, H-7), 7.51 (d, J = 8.7 Hz, 2H, Ho), 7.70 (s, 1H, Hβ), 8.17 (d, J = 8.9 Hz, 1H, H-8). 13C-NMR (DMSO-d6, 100 MHz) δ: 22.8 (C-4), 25.6 (C-3), 55.3 (OCH3), 57.1 (OCH3), 112.1 (C-7), 114.2, 126.3, 127.4, 131.6, 131.7 (C-8), 132.0, 135.6, 136.5 (Cβ), 139.0, 153.7, 159.9, 184.1 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 8.44; area: 98%. Anal. cal. for (C19H17NO5.0.5H2O): C, 65.51; H, 5.21, N, 4.02. Found: C, 65.94; H, 4.93, N, 4.23.
(E)-2-(3-Fluoro-4-methoxybenzylidene)-6-methoxy-5-nitro-3,4-dihydronaphthalen-1(2H)-one (8), following the general procedure A, 6-methoxy-5-nitro-1-tetralone (6) (150 mg, 0.68 mmol) reacted with 3-fluoro-4-methoxybenzaldehyde (128 mg, 0.83 mmol) in ethanol (3.0 mL). The precipitate was collected by filtration and washed with ethanol and water to give 208 mg (86% yield) of 8 as a yellow solid, which did not require further purification. Mp 189–190 °C. MS (ES, positive mode): m/z 358 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 2.80 (t, J = 6.5 Hz, 2H, H-4), 3.10 (t, J = 6.5 Hz, 2H, H-3), 3.89 (s, 3H, OCH3), 3.99 (s, 3H, OCH3), 7.25 (t, J = 8.8 Hz, 1H, Hm’), 7.36 (dd, J = 8.4, 2.4 Hz, 1H, Ho’), 7.40 (d, J = 8.9 Hz, 1H, H-7), 7.44 (dd, J = 12.8, 2.1 Hz, 1H, Ho), 7.66 (s, 1H, Hβ), 8.17 (d, J = 8.9 Hz, 1H, H-8). 13C-NMR (DMSO-d6, 100 MHz) δ: 22.7 (C-4), 25.5 (C-3), 56.1 (OCH3), 57.2 (OCH3), 112.1 (C-7), 113.8 (d, 3JC-F = 2.2 Hz, Cm’), 117.3 (d, 2JC-F = 18.5 Hz, Co), 126.1, 127.3 (d, 4JC-F = 3.2 Hz, Co’), 127.8 (d, 3JC-F = 6.9 Hz, C), 131.7 (C-8), 132.9, 135.2 (d, 4JC-F = 2.1 Hz, Cβ), 135.6, 139.0, 147.8 (d, 2JC-F = 10.6 Hz, C-OCH3), 151.1 (d, 1JC-F = 244.3 Hz, C-F), 153.7, 184.0 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 8.33; area: 97%. %. Anal. cal. for (C19H16FNO5): C, 63.86; H, 4.51, N, 3.92. Found: C, 63.72; H, 4.55, N, 3.78.
(E)-6-Methoxy-2-((6-methoxypyridin-3-yl)methylene)-5-Nitro-3,4-Dihydronaphthalen-1(2H)-one (9), following the general procedure A, 6-methoxy-5-nitro-1-tetralone (6) (100 mg, 0.45 mmol) reacted with 6-methoxy-3-pyridinecarboxaldehyde (76 mg, 0.55 mmol) in ethanol (3.0 mL). The precipitate was collected by filtration and washed with ethanol and water. The solid obtained was purified by flash column chromatography (CH2Cl2: MeOH 10:0.1) to give 125 mg (82% yield) of 9 as a yellow solid. Mp 176–178 °C. MS (ES, positive mode): m/z 341 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 2.81 (t, J = 6.5 Hz, 2H, H-4), 3.09 (t, J = 6.5 Hz, 2H, H-3), 3.90 (s, 3H, OCH3), 3.99 (s, 3H, OCH3), 6.92 (d, J = 8.7 Hz, 1H, Hm), 7.41 (d, J = 8.9 Hz, 1H, H-7), 7.69 (s, 1H, Hβ), 7.91 (dd, J = 8.7, 2.5 Hz, 1H, Ho), 8.18 (d, J = 8.9 Hz, 1H, H-8), 8.39 (d, J = 2.4 Hz, 1H, Ho’). 13C-NMR (DMSO-d6, 75 MHz) δ: 23.2 (C-4), 26.0 (C-3), 53.8 (OCH3), 57.5 (OCH3), 110.9, 112.5 (C-7), 124.9, 126.5, 132.1 (C-8), 133.4, 133.8, 136.1 (Cβ), 139.4, 140.6, 149.5, 154.2, 163.8, 184.2 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 7.61; area: 95%. %. Anal. cal. for (C18H16N2O5): C, 63.53; H, 4.74; N, 8.23. Found: C, 63.42; H, 4.89, N, 8.11.
(E)-6-Methoxy-5-Nitro-2-(3,4,5-trimethoxybenzylidene)-3,4-Dihydronaphthalen-1(2H)-one (10), following the general procedure A, 6-methoxy-5-nitro-1-tetralone (6) (250 mg, 1.13 mmol) reacted with 3,4,5-trimethoxybenzaldehyde (276 mg, 1.41 mmol) in ethanol (5.0 mL). The precipitate was collected by filtration and washed with ethanol and water. The solid obtained was purified by flash column chromatography (CH2Cl2) to give 347 mg (77% yield) of 10 as a yellow solid. Mp 204–205 °C. MS (ES, positive mode): m/z 400 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 2.81 (t, J = 6.5 Hz, 2H, H-4), 3.17 (t, J = 6.5 Hz, 2H, H-3), 3.71 (s, 3H, OCH3), 3.81 (s, 6H, OCH3), 3.99 (s, 3H, OCH3), 6.84 (s, 2H, Ho), 7.41 (d, J = 9.0 Hz, 1H, H-7), 7.70 (s, 1H, Hβ), 8.18 (d, J = 9.0 Hz, 1H, H-8). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.9 (C-4), 25.7 (C-3), 56.0 (OCH3), 57.2 (OCH3), 60.1 (OCH3), 107.7 (Co), 112.2 (C-7), 126.2, 130.4, 131.8 (C-8), 133.2, 135.7, 136.8 (Cβ), 138.3, 139.1, 152.8, 153.8, 184.1 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 8.00; area: 98%. Anal. cal. for (C21H21NO7): C, 63.15; H, 5.30, N, 3.51. Found: C, 63.08; H, 5.30, N, 3.64.

3.2.3. General Procedure for the Synthesis of N-Acetylpyrazolines 1114a,b (General Procedure B)

To a solution of the selected chalcone (710) (1.0 eq) in acetic acid (2.0 mL), hydrazine hydrate (10 eq) was added and the reaction was refluxed for 2 h. After completion of the reaction, the mixture was quenched by the addition of water (20 mL) and extracted with dichloromethane (3 × 20 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting products were purified by CCTLC in the Chromatothron as specified.
1-((3R,3aS)-rel-7-Methoxy-3-(4-methoxyphenyl)-6-nitro-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (11a) and 1-((3R,3aR)-rel-7-methoxy-3-(4-methoxyphenyl)-6-nitro-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (11b), following the general procedure B, compound 7 (150 mg, 0.45 mmol) reacted with hydrazine hydrate (220 μL, 4.5 mmol) in acetic acid (2.0 mL). The resulting products were purified by CCTLC in the Chromatothron (hexane:EtOAc 10:5). The fastest moving fractions provided 45 mg (26% yield) of 11a while the next fractions provided 11b. These later fractions required an additional purification by CCTLC in Chromatothron (CH2Cl2:MeOH, 10:0.1) to give 53 mg (30% yield) of 11b. Both compounds were obtained as pale yellow solids. Experimental data for 11a: Mp 107–109 °C. MS (ES, positive mode): m/z 396 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 1.91 (m, 1H, H-4), 2.09–2.36 (m, 4H, COCH3, H-4), 2.64 (m, 1H, H-5), 2.87 (m, 1H, H-5), 3.25 (ddd, J = 14.1, 9.7, 4.9 Hz, 1H, H-3a), 3.74 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 4.90 (d, J = 9.7 Hz, 1H, H-3), 6.89 (d, J = 8.7 Hz, 2H, Hm), 7.20 (d, J = 8.7 Hz, 2H, Ho), 7.35 (d, J = 8.9 Hz, 1H, H-8), 8.04 (d, J = 8.9 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 100 MHz) δ: 22.1 (COCH3), 23.7 (C-5), 25.8 (C-4), 53.8 (C-3a), 55.1 (OCH3), 57.0 (OCH3), 66.3 (C-3), 112.2 (C-8), 113.9 (Cm), 120.7, 127.2 (Co), 127.6 (C-9), 131.6, 133.8, 140.4, 151.3, 152.9, 158.3, 168.9 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 7.32; area: 98%. Anal. cal. for (C21H21N3O5): C, 63.79; H, 5.35, N, 10.63. Found: C,63.54; H, 5.38, N, 10.48. Experimental data for 11b: Mp 215–217 °C. MS (ES, positive mode): m/z 396 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.76 (m, 1H, H-4), 1.72 (m, 1H, H-4), 2.32 (s, 3H, COCH3), 2.58 (m, 1H, H-5), 2.79 (m, 1H, H-5), 3.66–3.77 (m, 4H, OCH3, H-3a), 3.93 (s, 3H, OCH3), 5.62 (d, J = 11.0 Hz, 1H, H-3), 6.85 (d, J = 8.6 Hz, 2H, Hm), 6.95 (d, J = 8.6 Hz, 2H, Ho), 7.35 (d, J = 8.9 Hz, 1H, H-8), 8.14 (d, J = 8.9 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 100 MHz) δ: 21.7 (COCH3), 22.6 (C-5), 23.7 (C-4), 46.9 (C-3a), 55.0 (OCH3), 57.0 (OCH3), 62.1 (C-3), 112.3 (C-8), 113.8 (Cm), 120.6, 127.1 (Co), 127.8 (C-9), 129.1, 131.7, 140.3, 151.5, 153.4, 158.5, 166.9 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 5.58; area: 98%. Anal. cal. for (C21H21N3O5): C, 63.79; H, 5.35, N, 10.63. Found: C,63.60; H, 5.40, N, 10.49.
1-((3R,3aS)-rel-3-(3-Fluoro-4-methoxyphenyl)-7-methoxy-6-nitro-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (12a) and 1-((3R,3aR)-rel-3-(3-fluoro-4-methoxyphenyl)-7-methoxy-6-nitro-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (12b), following the general procedure B, compound 8 (200 mg, 0.56 mmol) reacted with hydrazine hydrate (280 μL, 5.8 mmol) in acetic acid (2.0 mL). The resulting products were purified by CCTLC in the Chromatothron (hexane:EtOAc 10:5). The fastest moving fractions provided 93 mg (40% yield) of 12a, while the next fractions provided 12b, which required an additional purification by CCTLC in Chromatothron (CH2Cl2:MeOH, 10:0.1) to give 80 mg (34% yield) of 12b. Both compounds were obtained as pale yellow solids. Experimental data for 12a: Mp 209–210 °C. MS (ES, positive mode): m/z 414 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 1.90 (m, 1H, H-4), 2.13–2.34 (m, 4H, COCH3, H-4), 2.64 (m, 1H, H-5), 2.87 (m, 1H, H-5), 3.28 (ddd, J = 14.8, 9.8, 4.9 Hz, 1H, H-3a), 3.82 (s, 3H, OCH3), 3.94 (s, 3H, OCH3), 4.91 (d, J = 9.8 Hz, 1H, H-3), 7.07 (dd, J = 8.5, 2.1 Hz, 1H, Ho’), 7.10–7.17 (m, 2H, Ho, Hm’), 7.35 (d, J = 8.9 Hz, 1H, H-8), 8.04 (d, J = 8.9 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.1 (COCH3), 23.6 (C-5), 25.7 (C-4), 53.6 (C-3a), 56.0 (OCH3), 57.0 (OCH3), 65.9 (C-3), 112.2 (C-8), 113.7 (d, 2JC-F = 19.0 Hz, Co), 113.8 (d, 3JC-F = 1.6 Hz, Cm’), 120.6, 122.3 (d, 4JC-F = 3.5 Hz, Co’), 127.7 (C-9), 131.6, 134.7 (d, 3JC-F = 6.1 Hz, C-C3), 140.4, 146.1 (d, 2JC-F = 10.5 Hz, C-OCH3), 151.3, 151.4 (d, 1JC-F = 243.8 Hz, C-F), 153.0, 169.0 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 7.43; area: 97%. Anal. cal. for (C21H20FN3O5): C, 61.01; H, 4.88, N, 10.16. Found: C, 60.85; H, 4.93, N, 10.04. Experimental data for 12b: Mp 127–129 °C. MS (ES, positive mode): m/z 414 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.79 (m, 1H, H-4), 1.77 (m, 1H, H-4), 2.33 (s, 3H, COCH3), 2.58 (m, 1H, H-5), 2.79 (m, 1H, H-5), 3.72 (ddd, J = 13.7, 11.0, 4.9 Hz, 1H, H-3a), 3.79 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 5.63 (d, J = 11.0 Hz, 1H, H-3), 6.80 (d, J = 8.3 Hz, 1H, Ho’), 6.90 (dd, J = 12.2, 2.1 Hz, 1H, Ho), 7.09 (t, J = 8.7 Hz, 1H, Hm’), 7.35 (d, J = 9.0 Hz, 1H, H-8), 8.14 (d, J = 9.0 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 21.6 (COCH3), 22.6 (C-5), 23.8 (C-4), 46.9 (C-3a), 56.0 (OCH3), 57.0 (OCH3), 61.6 (C-3), 112.3 (C-8), 113.7 (d, 2JC-F = 18.9 Hz, Co), 113.9 (d, 3JC-F = 0.6 Hz, Cm’), 120.5, 122.0 (d, 4JC-F = 3.2 Hz, Co’), 127.9 (C-9), 130.1 (d, 3JC-F = 5.5 Hz, C-C3), 131.7, 140.3, 146.2 (d, 2JC-F = 10.5 Hz, C-OCH3), 151.3 (d, 1JC-F = 244.1 Hz, C-F), 151.6, 153.5, 167.0 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 6.93; area: 98%. Anal. cal. for (C21H20FN3O5.0.25H20): C, 60.36; H, 4.94, N, 10.06. Found: C, 60.32; H, 4.88, N, 10.08.
1-((3R,3aS)-rel-7-Methoxy-3-(6-methoxypyridin-3-Yl)-6-nitro-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (13a) and 1-((3R,3aR)-rel-7-methoxy-3-(6-methoxypyridin-3-Yl)-6-nitro-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (13b), following the general procedure B, compound 9 (234 mg, 0.69 mmol) reacted with hydrazine hydrate (340 μL, 7.0 mmol) in acetic acid (2.0 mL). The resulting products were purified by CCTLC in the Chromatothron (hexane:EtOAc 10:5). The fastest moving fractions provided 104 mg (38% yield) of 13a, while the next fractions provided 13b, which required an additional purification by CCTLC in Chromatothron (CH2Cl2:MeOH, 10:0.2) to give 90 mg (33% yield) of 13b. Both compounds were obtained as pale yellow solids. Experimental data for 13a: Mp 208–210 °C. MS (ES, positive mode): m/z 397 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 1.92 (m, 1H, H-4), 2.17–2.30 (m, 4H, COCH3, H-4), 2.65 (m, 1H, H-5), 2.87 (m, 1H, H-5), 3.36 (ddd, J = 14.4, 10.0, 4.9 Hz, 1H, H-3a), 3.84 (s, 3H, OCH3), 3.94 (s, 3H, OCH3), 4.96 (d, J = 10.0 Hz, 1H, H-3), 6.79 (d, J = 8.6 Hz, 1H, Hm), 7.36 (d, J = 8.9 Hz, 1H, H-8), 7.63 (dd, J = 8.6, 2.5 Hz, 1H, Ho), 8.05 (d, J = 8.9 Hz, 1H, H-9), 8.12 (d, J = 2.5 Hz, 1H, Ho’). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.0 (COCH3), 23.7 (C-5), 25.5 (C-4), 53.1 (C-3a), 53.2 (OCH3), 57.0 (OCH3), 64.1 (C-3), 110.4, 112.3 (C-8), 120.6, 127.7 (C-9), 130.1, 131.7, 137.1, 140.4, 145.0, 151.4, 153.1, 162.8, 169.1 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 6.41; area: 96%. Anal. cal. for (C20H20N4O5): C, 60.60; H, 5.09, N, 14.13. Found: C, 60.42; H, 5.15, N, 14.02. Experimental data for 13b: Mp 123–125 °C. MS (ES, positive mode): m/z 397 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.86 (m, 1H, H-4), 1.78 (m, 1H, H-4), 2.32 (s, 3H, COCH3), 2.60 (m, 1H, H-5), 2.81 (m, 1H, H-5), 3.76 (ddd, J = 13.6, 11.0, 4.9 Hz, 1H, H-3a), 3.81 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 5.68 (d, J = 11.0 Hz, 1H, H-3), 6.74 (d, J = 8.6 Hz, 1H, Hm), 7.32 (dd, J = 8.6, 2.6 Hz, 1H, Ho), 7.36 (d, J = 9.0 Hz, 1H, H-8), 7.92 (d, J = 2.5 Hz, 1H, Ho’), 8.14 (d, J = 9.0 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 21.6 (COCH3), 22.7 (C-5), 23.8 (C-4), 46.7 (C-3a), 53.1 (OCH3), 57.0 (OCH3), 59.9 (C-3), 110.4, 112.4 (C-8), 120.5, 125.8, 127.9 (C-9), 131.7, 137.1, 140.3, 144.6, 151.6, 153.6, 163.0, 167.0 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 5.98; area: 96%. Anal. cal. for (C20H20N4O5): C, 60.60; H, 5.09, N, 14.13. Found: C, 60.49; H, 5.18, N, 14.05.
1-((3R,3aS)-rel-7-Methoxy-6-nitro-3-(3,4,5-trimethoxyphenyl)-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (14a) and 1-((3R,3aR)-rel-7-methoxy-6-nitro-3-(3,4,5-trimethoxyphenyl)-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (14b), following the general procedure B, compound 10 (260 mg, 0.65 mmol) reacted with hydrazine hydrate (320 μL, 6.6 mmol) in acetic acid (2.0 mL). The resulting products were purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc 20:1). The fastest moving fractions provided 55 mg (19 % yield) of 14a, while the next fractions provided 108 mg (36% yield) of a mixture 14a and 14b, and the final fractions provided 32 mg (11 % yield) of 14b. The mixture of diasteromers 14a and 14b, was submitted to two additional purification by CCTLC in Chromatothron using hexane:EtOAc (10:5) and CH2Cl2:MeOH (10:0.1), however both compounds were obtained in almost all collect fractions. The mixture 14a and 14b was used in the next step. Data for 14a and 14b were obtained from fractions containing a single diasteromer. Experimental data for 14a: Mp 282–283 °C. MS (ES, positive mode): m/z 456 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 1.92 (m, 1H, H-4), 2.20–2.30 (m, 4H, COCH3, H-4), 2.65 (m, 1H, H-5), 2.87 (m, 1H, H-5), 3.29 (ddd, J = 13.2, 9.9, 4.9 Hz, 1H, H-3a), 3.64 (s, 3H, OCH3), 3.76 (s, 6H, OCH3), 3.94 (s, 3H, OCH3), 4.88 (d, J = 9.9 Hz, 1H, H-3), 6.57 (s, 2H, Ho), 7.35 (d, J = 8.9 Hz, 1H, H-8), 8.06 (d, J = 8.9 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.1 (COCH3), 23.6 (C-5), 25.8 (C-4), 53.7 (C-3a), 55.9 (OCH3), 57.0 (OCH3), 59.9 (OCH3), 67.1 (C-3), 103.1 (C-8), 112.2, 120.7, 127.7 (C-9), 131.7, 136.3, 137.7, 140.4, 151.3, 152.9, 153.0, 169.1 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 6.66; area: 98%. Anal. cal. for (C23H25N3O7): C, 60.65; H, 5.53, N, 9.23. Found: C, 60.35; H, 5.51, N, 8.99. Experimental data for 14b: Mp 270–272 °C. MS (ES, positive mode): m/z 456 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.84 (m, 1H, H-4), 1.83 (m, 1H, H-4), 2.35 (s, 3H, COCH3), 2.61 (m, 1H, H-5), 2.80 (m, 1H, H-5), 3.61 (s, 3H, OCH3), 3.65–3.81 (m, 7H, H-3a, OCH3), 3.93 (s, 3H, OCH3), 5.62 (d, J = 11.1 Hz, 1H, H-3), 6.31 (s, 2H, Ho), 7.34 (d, J = 8.9 Hz, 1H, H-8), 8.14 (d, J = 8.9 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 21.6 (COCH3), 22.5 (C-5), 23.8 (C-4), 46.9 (C-3a), 55.8 (OCH3), 57.0 (OCH3), 59.9 (OCH3), 62.6 (C-3), 103.0, 112.3 (C-8), 120.6, 127.9, 131.8, 133.0, 136.5, 140.3, 151.5, 152.9, 153.7, 167.1 (C=O). Analytical HPLC (gradient 30-95% acetonitrile in 10 min): TR: 6.18; area: 96%. Anal. cal. for (C23H25N3O7): C, 60.65; H, 5.53, N, 9.23. Found: C, 60.39; H, 5.62, N, 9.12.

3.2.4. General Procedure for the Synthesis of Pyrazolines Bearing 6-Amino-7-Methoxy-3,3a,4,5-Tetrahydro-2H-Benzo[g]indazole moieties 1518a,b (General Procedure C)

To a suspension containing the appropriate nitro compound (1.0 eq) and iron powder (10 eq) in ethanol-water (2:1, 3.0 mL), one drop of 37% HCl was added. The reaction mixture was heated to reflux for 1 h. EtOAc (10 mL) was added to the mixture, washed with water and the aqueous phase was further extracted with EtOAc (3 × 10 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure. The residue was purified by CCTLC in the Chromatothron as specified.
1-((3R,3aS)-rel-6-Amino-7-methoxy-3-(4-methoxyphenyl)-3,3a,4,5-tetrahydro-2H-tenzo[g]indazol-2-yl)ethan-1-one (15a), following the general procedure C, compound 11a (60 mg, 0.15 mmol) reacted with iron powder (88 mg, 1.6 mmol) in EtOH:H2O (2:1, 3.0 mL). The crude product was purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc, 10:2) to provide 40 mg (73% yield) of 15a, as a beige solid. Mp 219–220 °C. MS (ES, positive mode): m/z 366 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 1.83 (m, 1H, H-4), 2.15–2.27 (m, 4H, COCH3, H-4), 2.45 (m, 1H, H-5), 2.74 (m, 1H, H-5), 3.05 (ddd, J = 13.8, 9.2, 4.8 Hz, 1H, H-3a), 3.74 (s, 3H, OCH3), 3.82 (s, 3H, OCH3), 4.61 (br s, 2H, NH2), 4.83 (d, J = 9.2 Hz, 1H, H-3), 6.85 (d, J = 8.5 Hz, 1H, H-8), 6.89 (d, J = 8.6 Hz, 2H, Hm), 7.18 (d, J = 8.6 Hz, 2H, Ho), 7.19 (d, J = 8.5 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.1 (COCH3), 23.8 (C-5), 26.8 (C-4), 54.7 (C-3a), 55.1 (OCH3), 55.6 (OCH3), 66.0 (C-3), 108.8 (C-8), 112.8 (C-9), 113.9 (Cm), 119.9, 123.4, 127.0 (Co), 134.4, 134.7, 147.5, 155.6, 158.2, 168.4 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 5.12; area: 97%. Anal. cal. for (C21H23N3O3): C, 69.02; H, 6.34, N, 11.50. Found: C, 68.70; H, 6.47, N, 11.21.
1-((3R,3aR)-rel-6-Amino-7-methoxy-3-(4-methoxyphenyl)-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (15b), following the general procedure C, compound 11b (71 mg, 0.18 mmol) reacted with iron powder (107 mg, 1.9 mmol) in EtOH:H2O (2:1, 3.0 mL). The crude product was purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc, 10:2) to provide 49 mg (75% yield) of 15b, as a white solid. Mp 217–219 °C. MS (ES, positive mode): m/z 366 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.72 (m, 1H, H-4), 1.72 (m, 1H, H-4), 2.28 (s, 3H, COCH3), 2.40 (m, 1H, H-5), 2.68 (m, 1H, H-5), 3.57 (ddd, J = 15.2, 10.8, 4.7 Hz, 1H, H-3a), 3.71 (s, 3H, OCH3), 3.81 (s, 3H, OCH3), 4.58 (br s, 2H, NH2), 5.55 (d, J = 10.8 Hz, 1H, H-3), 6.82–6.88 (m, 3H, Hm, H-8), 6.96 (d, J = 8.3 Hz, 2H, Ho), 7.31 (d, J = 8.4 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 21.7 (COCH3), 23.3 (C-5), 23.6 (C-4), 47.3 (C-3a), 55.0 (OCH3), 55.6 (OCH3), 61.8 (C-3), 108.9 (C-8), 113.1 (C-9), 113.7 (Cm), 119.8, 123.7, 127.1 (Co), 129.6, 134.7, 147.7, 156.2, 158.3, 166.4 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 4.22; area: 98%. Anal. cal. for (C21H23N3O3): C, 69.02; H, 6.34, N, 11.50. Found: C, 68.64; H, 6.53, N, 11.04.
1-((3R,3aS)-rel-6-Amino-3-(3-fluoro-4-methoxyphenyl)-7-methoxy-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (16a), following the general procedure C, compound 12a (69 mg, 0.17 mmol) reacted with iron powder (102 mg, 1.8 mmol) in EtOH:H2O (2:1, 3.0 mL). The crude product was purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc, 10:2) to provide 43 mg (67% yield) of 16a, as a white solid. Mp 154–156 °C. MS (ES, positive mode): m/z 384 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 1.82 (m, 1H, H-4), 2.15–2.35 (m, 4H, COCH3, H-4), 2.45 (m, 1H, H-5), 2.74 (m, 1H, H-5), 3.09 (ddd, J = 13.9, 9.4, 4.8 Hz, 1H, H-3a), 3.82 (s, 6H, OCH3), 4.61 (br s, 2H, NH2), 4.84 (d, J = 9.4 Hz, 1H, H-3), 6.85 (d, J = 8.5 Hz, 1H, H-8), 7.05 (dd, J = 8.5, 2.1 Hz, 1H, Ho’), 7.09–7.14 (m, 2H, Ho, Hm’), 7.20 (d, J = 8.4 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.1 (COCH3), 23.8 (C-5), 26.7 (C-4), 54.4 (C-3a), 55.6 (OCH3), 56.0 (OCH3), 65.6 (C-3), 108.8 (C-8), 112.8 (C-9), 113.5 (d, 2JC-F = 18.7 Hz, Co), 113.9 (d, 3JC-F = 1.2 Hz, Cm’), 119.8, 122.0 (d, 4JC-F = 3.4 Hz, Co’), 123.5, 134.8, 135.2 (d, 3JC-F = 6.0 Hz, C-C3), 146.0 (d, 2JC-F = 10.6 Hz, C-OCH3), 147.6, 151.4 (d, 1JC-F = 243.6 Hz, C-F), 155.7, 168.6 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 5.13; area: 97%. Anal. cal. for (C21H22FN3O3): C, 65.78; H, 5.78, N, 10.96. Found: C, 65.82; H, 5.89, N, 10.78.
1-((3R,3aR)-rel-6-Amino-3-(3-fluoro-4-methoxyphenyl)-7-methoxy-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (16b), following the general procedure C, compound 12b (50 mg, 0.12 mmol) reacted with iron powder (67 mg, 1.2 mmol) in EtOH:H2O (2:1, 3.0 mL). The crude product was purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc, 10:2) to provide 27 mg (59% yield) of 16b, as a white solid. Mp 197–199 °C. MS (ES, positive mode): m/z 384 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.73 (m, 1H, H-4), 1.77 (m, 1H, H-4), 2.29 (s, 3H, COCH3), 2.42 (m, 1H, H-5), 2.70 (m, 1H, H-5), 3.59 (ddd, J = 13.8, 10.9, 4.9 Hz, 1H, H-3a), 3.79 (s, 3H, OCH3), 3.81 (s, 3H, OCH3), 4.59 (br s, 2H, NH2), 5.56 (d, J = 10.9 Hz, 1H, H-3), 6.80 (d, J = 8.5 Hz, 1H, Ho), 6.85 (d, J = 8.6 Hz, 1H, H-8), 6.89 (dd, J = 8.5, 2.1 Hz, 1H, Ho’), 7.09 (t, J = 8.7 Hz, 1H, Hm’), 7.31 (d, J = 8.6 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 21.6 (COCH3), 23.2 (C-5), 23.6 (C-4), 47.3 (C-3a), 55.6 (OCH3), 56.0 (OCH3), 61.3 (C-3), 108.9 (C-8), 113.1 (C-9), 113.7 (d, 2JC-F = 18.9 Hz, Co), 113.8 (Cm’), 119.7, 122.0 (d, 4JC-F = 3.1 Hz, Co’), 123.6, 130.7 (d, 3JC-F = 5.5 Hz, C-C3), 134.7, 146.1 (d, 2JC-F = 10.5 Hz, C-OCH3), 147.7, 151.3 (d, 1JC-F = 244.2 Hz, C-F), 156.2, 166.5 (C=O). Analytical HPLC (gradient 30-95% acetonitrile in 10 min): TR: 4.35; area: 95%). Anal. cal. for (C21H22FN3O3): C, 65.78; H, 5.78, N, 10.96. Found: C, 65.87; H, 5.91, N, 10.82.
1-((3R,3aS)-rel-6-Amino-7-methoxy-3-(6-methoxypyridin-3-yl)-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (17a), following the general procedure C, compound 13a (63 mg, 0.16 mmol) reacted with iron powder (90 mg, 1.6 mmol) in EtOH:H2O (2:1, 3 mL). The crude product was purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc, 10:4) to provide 30 mg (52% yield) of 17a, as a white solid. Mp 185–187 °C. MS (ES, positive mode): m/z 367 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz δ 1.83 (m, 1H, H-4), 2.16–2.31 (m, 4H, COCH3, H-4), 2.45 (m, 1H, H-5), 2.75 (m, 1H, H-5), 3.16 (ddd, J = 13.3, 9.6, 4.8 Hz, 1H, H-3), 3.82 (s, 3H, OCH3), 3.84 (s, 3H, OCH3), 4.62 (br s, 2H, NH2), 4.88 (d, J = 9.6 Hz, 1H, H-3a), 6.78 (d, J = 8.6 Hz, 1H, Hm), 6.85 (d, J = 8.5 Hz, 1H, H-8), 7.21 (d, J = 8.5 Hz, 1H, H-9), 7.60 (dd, J = 8.6, 2.5 Hz, 1H, Ho), 8.11 (d, J = 2.5 Hz, 1H, Ho’). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.1 (COCH3), 23.8 (C-5), 26.5 (C-4), 53.2 (OCH3), 53.9 (C-3a), 55.6 (OCH3), 63.9 (C-3), 108.8 (C-8), 110.4 (Cm), 112.9 (C-9), 119.7, 123.5, 130.6, 134.8, 137.0 (Co), 144.8 (Co’), 147.6, 155.8, 162.7, 168.7 (C=O). Analytical HPLC (gradient 30-95% acetonitrile in 10 min): TR: 2.67; area: 98%. Anal. cal. for (C20H22N4O3): C, 65.56; H, 6.05; N, 15.29. Found: C, 65.13; H, 6.09; N, 15.00.
1-((3R,3aR)-rel-6-Amino-7-methoxy-3-(6-methoxypyridin-3-yl)-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (17b), following the general procedure C, compound 13b (65 mg, 0.16 mmol) reacted with iron powder (97 mg, 1.7 mmol) in EtOH:H2O (2:1, 3.0 mL). The crude product was purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc, 10:4) to provide 29 mg (50% yield) of 17b, as a white solid. Mp 202–204 °C. MS (ES, positive mode): m/z 367 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.80 (m, 1H, H-4), 1.77 (m, 1H, H-4), 2.28 (s, 3H, COCH3), 2.43 (m, 1H, H-5), 2.71 (m, 1H, H-5), 3.63 (ddd, J = 15.0, 11.0, 4.7 Hz, 1H, H-3a), 3.81 (s, 3H, OCH3), 3.82 (s, 3H, OCH3), 4.60 (br s, 2H, NH2), 5.61 (d, J = 11.0 Hz, 1H), 6.74 (d, J = 8.5 Hz, 1H, Hm), 6.86 (d, J = 8.5 Hz, 1H, H-8), 7.26–7.35 (m, 2H, H-9, Ho), 7.91 (d, J = 2.5 Hz, 1H, Ho’). 13C-NMR (DMSO-d6, 75 MHz) δ: 21.7 (COCH3), 23.5 (C-5), 23.7 (C-4), 47.3 (C-3a), 53.2 (OCH3), 55.7 (OCH3), 59.6 (C-3), 108.9 (C-8), 110.3 (Cm), 113.2 (C-9), 119.7, 123.7, 126.3, 134.7, 137.2 (Co), 144.6 (Co’), 147.8, 156.4, 162.9, 166.7 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 2.08; area: 96%. Anal. cal. for (C20H22N4O3): C, 65.56; H, 6.05; N, 15.29. Found: C, 65.24; H, 6.12; N, 15.07.
1-((3R,3aS)-rel-6-Amino-7-methoxy-3-(3,4,5-trimethoxyphenyl)-3,3a,4,5-tetrahydro-2H-benzo[g]indazol-2-yl)ethan-1-one (18a) and 1-((3R,3aR)-rel-6-amino-7-methoxy-3-(3,4,5-trimethoxy- phenyl)-3,3a,4,5-tetrahydro-2h-benzo[g]indazol-2-yl)Ethan-1-One (18b), following the general procedure C, a mixture of compounds 14a and 14b (74 mg, 0.16 mmol) reacted with iron powder (100 mg, 1.8 mmol) in EtOH:H2O (2:1, 3.0 mL). The crude product was purified by CCTLC in the Chromatothron (CH2Cl2:EtOAc, 10:2). The fastest moving fractions afforded 17 mg (25% yield) of 18a, while the slowest moving fractions provided 30 mg (43% yield) of 18b. Both compounds were obtained as white solids. Experimental data for 18a: Mp 225–227 °C. MS (ES, positive mode): m/z 426 [M + H]+. 1H-NMR (DMSO-d6, 400 MHz) δ: 1.83 (m, 1H, H-4), 2.23–2.32 (m, 4H, COCH3, H-4), 2.47 (m, 1H, H-5), 2.75 (m, 1H, H-5), 3.11 (ddd, J = 14.0, 9.5, 4.9 Hz, 1H, H-3a), 3.64 (s, 3H, OCH3), 3.76 (s, 6H, OCH3), 3.82 (s, 3H, OCH3), 4.62 (br s, 2H, NH2), 4.80 (d, J = 9.5 Hz, 1H, H-3), 6.54 (s, 2H, Ho), 6.85 (d, J = 8.5 Hz, 1H, H-8), 7.21 (d, J = 8.5 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 22.1 (COCH3), 23.8 (C-5), 26.8 (C-4), 54.5 (C-3a), 55.6 (OCH3), 55.8 (OCH3), 59.9 (OCH3), 66.8 (C-3), 102.8 (Co), 108.8 (C-8), 112.8 (C-9), 119.8, 123.5, 134.8, 136.2, 138.2, 147.5, 153.0, 155.6, 168.7 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 4.67; area: 95%. Anal. cal. for (C23H27N3O5): C, 64.93; H, 6.40; N, 9.88. Found: C, 64.81; H, 6.55; N, 9.85. Experimental data for 18b: Mp 181–183 °C. MS (ES, positive mode): m/z 426 (M+H)+. 1H-NMR (DMSO-d6, 400 MHz) δ: 0.78 (m, 1H, H-4), 1.83 (m, 1H, H-4), 2.32 (s, 3H, COCH3), 2.41 (m, 1H, H-5), 2.70 (m, 1H, H-5), 3.55 – 3.67 (m, 4H, H-3a, OCH3), 3.71 (s, 6H, OCH3), 3.82 (s, 3H, OCH3), 4.59 (br s, 2H, NH2), 5.56 (d, J = 11.0 Hz, 1H, H-3), 6.32 (s, 2H, Ho), 6.85 (d, J = 8.6 Hz, 1H, H-8), 7.32 (d, J = 8.6 Hz, 1H, H-9). 13C-NMR (DMSO-d6, 75 MHz) δ: 21.6 (COCH3), 23.1 (C-5), 23.7 (C-4), 47.3 (C-3a), 55.6 (OCH3), 55.8 (OCH3), 59.9 (OCH3), 62.3 (C-3), 103.1 (Co), 108.8 (C-8), 113.1, (C-9) 119.8, 123.7, 133.5, 134.7, 136.4, 147.7, 152.8, 156.4, 166.6 (C=O). Analytical HPLC (gradient 30–95% acetonitrile in 10 min): TR: 3.62; area: 97%. Anal. cal. for (C23H27N3O5): C, 64.93; H, 6.40; N, 9.88. Found: C, 64.79; H, 6.52; N, 9.81.

3.3. Antiproliferative Activity

The tumor cell lines were acquired from the American Type Culture Collection (ATCC, Manassas, VA, USA), except for the DND-41 cell line, which was purchased from the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ Leibniz-Institut, Germany), and the Hap-1 cell line was ordered from Horizon Discovery (Horizon Discovery Group, Waterbeach, UK). All cell lines were cultured as recommended by the suppliers. Media were purchased from Gibco Life Technologies (Waltham, MA, USA), and supplemented with 10% fetal bovine serum (HyClone, GE Healthcare Life Sciences, Chicago, IL, USA). Adherent cell lines HCT-116, NCI-H460, Hap-1 and Capan-1 cells were seeded at a density between 500 and 1500 cells per well, in 384-well, black walled, clear-bottomed tissue culture plates (Greiner Bio-One, Kremsmünster, Austria). After overnight incubation, cells were treated with the test compounds at seven different concentrations ranging from 100 to 6.4 × 10−3 µM. Suspension cell lines HL-60, K-562 and DND-41 were seeded at densities ranging from 2500 to 5500 cells per well in 384-well, black walled, clear-bottomed tissue culture plates containing the test compounds at the same seven concentration points.
The plates were incubated and monitored at 37 °C for 72 h in an IncuCyte® (Essen BioScience Inc., Ann Arbor, MI, USA) for real-time imaging of cell proliferation. Images were taken every 3 h, with one field imaged per well under 10× magnification. Area under the curve (AUC) values were calculated and used to determine the IC50 values.

3.4. Antibacterial Activity

The nitro compounds were tested to determine the antibacterial activity against gram-negative and gram-positive bacteria. Wild-type and multi-resistant strains were included as follows: methicillin-susceptible Staphylococcus aureus ATCC 25923 (MSSA), methicillin-resistant Staphylococcus aureus ATCC 43300 (MRSA), vancomycin-intermediate Staphylococcus aureus (VISA), Escherichia coli ATCC 25922, carbapenemase–positive Klebsiella pneumoniae BAA 1705, Klebsiella pneumoniae ATCC 700603 (extended spectrum beta lactamase, ESBL positive), Pseudomonas aeruginosa ATCC 27853 and Neisseria gonorrhoeae ATCC 49226. Stock solutions (100 mg/mL) of the compounds were prepared in dimethyl sulfoxide (DMSO) and diluted to a final screening concentration of 1 mg/mL. An initial screening of bacterial inhibition was performed by agar diffusion method. Briefly, sterile Mueller Hinton agar (MHA, BBL) was prepared in Petri dishes and inoculated with a bacterial suspension prepared in trypticase soy broth and adjusted to 1.5 × 108 colony forming unit CFU/mL (i.e., 0.08–0.1 OD at 600 nm) [33]. Wells (6 mm in diameter) were punched in the agar and 10 µL of each compound (stock solution) was filled into each well. Dimethyl sulfoxide and trypticase soy broth were included as negative controls (i.e., no inhibition of bacterial growth). Gentamicin and tetracycline (Sigma-Aldrich) were included as positive controls of growth inhibition. Compounds showing growth inhibition were tested at least twice before being selected for microdilution testing. For N. gonorrhoeae the agar diffusion method was also used in the screening process with some modifications. For this method, 200 µL of a bacterial suspension (1.5 × 108 CFU/mL) was inoculated in gonococcal (GC) agar (BBL) supplemented with 1% isovitalex (BBL), then the compounds were added to the wells as above mentioned and incubated at 35–36.5 °C in 5% CO2 atmosphere for 48 h. Penicillin, ceftriaxone and ciprofloxacin (BBL) were used as controls [34].

3.4.1. Microdilution Test

Minimum inhibitory concentration (MIC) was determined in those compounds with reproducible bacterial growth inhibition at the screening. Bacterial suspensions were adjusted with Mueller Hinton broth (MHB) to a concentration of 5 × 105–8 × 105 [33]. Stock solutions of the newly synthesized compounds were diluted in MHB containing 5% DMSO and 0.1% Tween 80 [35] and added to 90 µL of the bacterial inoculum. The microplates were incubated for 24 h at 35–37 °C. MICs were defined as the lowest concentration with visible inhibition of bacterial growth [33] and/or detection using resazurin (1 mg/mL) using a microplate spectrophotometer (Cytation 3M, Biotek, Winooski, VT, USA). Gentamicin (Sigma-Aldrich, St. Louis, MO, USA) and tetracycline were included as controls of growth inhibition; MHB and DMSO were used as a negative control. Experiments were performed in duplicate and replicated at least three times.
For N. gonorrhoeae, those compounds with visible growth inhibition in the screening test were further tested for MIC on agar plates as described by the Centers for Disease Control and Prevention [36] and the Clinical and Laboratory Standards Institute [34] with modifications. Briefly, GC agar supplemented with 1% isovitalex was prepared to contain increasing concentrations of the compounds and inoculated with 5 µL of the bacterial suspension (i.e., 1 × 104 CFU) [36]. The lowest concentration of the compound that inhibited bacterial growth was determined as the MIC. Bacterial growth was examined and verified using the oxidase test. Experiments were performed in duplicate and replicated at least three times.

3.4.2. Hemolytic Activity

The ability to induce hemolysis was evaluated to compounds that showed antibacterial activity following the method of cytotoxicity by spectrophotometry. The method was adapted from Conceição et al. [37] with modifications. Briefly, 240 μL of Human Red blood cells (huRBC) adjusted to 5% hematocrit in phosphate buffer saline (PBS) were placed into each well of 96-well plate and subsequently exposed to 200 µg/mL of the selected compounds (i.e., 10 μL of 5 mg/mL working solution of each compound in MHB with 5% DMSO 0.1% Tween-80). As positive control for hemolytic activity 10 μL SDS 1% was added. For a negative control only medium with no chemicals was added to huRBC. Free hemoglobin was measured after 24 h incubation at 37 °C by spectrophotometry (420 nm Cytation 3M, Biotek, Winooski, VT, USA). Non-specific absorbance was subtracted from a blank. Determinations were done by triplicate in at least two independent experiments.

4. Conclusions

A series of new benzo[g]indazole compounds 1118a,b incorporating 6-nitro and 6-amino groups were synthesized by cyclocondensation of 2-benzylidene-1-tetralones 710 with hydrazine hydrate in presence of acetic acid, resulting in the formation of cis (3R,3aR-rel) and trans (3R,3aS-rel) diastereoisomeric pyrazoline derivatives. The stereochemical assignment of the obtained diasteroisomers was performed based on experimental NMR data and a convenient criterion concerning the chemical shift of the H-4 protons has been proposed for the assignment of both isomers. The antiproliferative evaluation against a panel of tumor cell lines revealed that the 6-nitro benzo[g]indazoles 11a, 11b, 12a and 12b display IC50 values between 5–15 µM against NCI-H460 (lung carcinoma) while they were less active against other cell types. The in vitro screening for antibacterial activity showed that compounds 12a and 13b exhibited antigonococcal activity by inhibiting bacterial growth at MIC values of 250 and 62.5 μg/mL, respectively. None of the active compounds (12a and 13b) showed hemolytic activity suggesting low membrane interactions and toxicity.

Supplementary Materials

The following are available online. NMR spectra of representative compounds in each series. Additional biological methods: tubulin staining, gamma H2A.X immunofluorescence staining and DNA intercalation assay.

Author Contributions

Conceptualization: E.-M.P., M.-J.C., B.I. and M.-J.P.-P.; Funding acquisition: M.-J.C., B.I. and M.-J.P.-P.; Investigation: V.C., M.d.P.C., E.-M.P., L.P. and M.-J.P.-P.; Project administration: D.D., M.-J.C., B.I. and M.-J.P.-P.; Supervision: B.I. and M.-J.P.-P.; Visualization: V.C., E.-M.P.; Writing original draft: V.C., M.-J.P.-P.; Writing—review and editing: V.C., M.d.P.C., E.-M.P., D.D., M.-J.C., B.I. and M.-J.P.-P.

Funding

V.C.: M.P.C. and B.I. thank the Universidad del Valle (CIAM-2017), COLCIENCIAS (110680864255) and the Science, Technology and Innovation Fund-General Royalties System (FCTeI-SGR) under contract BPIN 2013000100007, Colombia. M.-J.P.-P., E.-M.P. and M.-J.C. acknowledge the financial support of SAF2015-64629-C2-1-R and CSIC-PIE-201680E079.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gaikwad, D.D.; Chapolikar, A.D.; Devkate, C.G.; Warad, K.D.; Tayade, A.P.; Pawar, R.P.; Domb, A.J. Synthesis of Indazole Motifs and Their Medicinal Importance: An Overview. Eur. J. Med. Chem. 2015, 90, 707–731. [Google Scholar] [CrossRef] [PubMed]
  2. Denya, I.; Malan, S.F.; Joubert, J. Indazole Derivatives and Their Therapeutic Applications: A Patent Review (2013–2017). Expert Opin. Ther. Pat. 2018, 28, 441–453. [Google Scholar] [CrossRef] [PubMed]
  3. Gao, M.; Lola, C.M.; Wang, M.; Miller, K.D.; Sledge, G.W.; Hutchins, G.D.; Zheng, Q.H. Synthesis of Carbon-11-Labeled Tricyclic Necroptosis Inhibitors as New Potential PET Agents for Imaging of Tumor Necrosis Factor α (TNF-α). Appl. Radiat. Isot. 2010, 68, 1950–1958. [Google Scholar] [CrossRef] [PubMed]
  4. Choi, S.; Keys, H.; Staples, R.J.; Yuan, J.; Degterev, A.; Cuny, G.D. Optimization of Tricyclic Nec-3 Necroptosis Inhibitors for in Vitro Liver Microsomal Stability. Bioorg. Med. Chem. Lett. 2012, 22, 5685–5688. [Google Scholar] [CrossRef] [PubMed]
  5. Collins, I.; Rowley, M.; Davey, W.B.; Emms, F.; Marwood, R.; Patel, S.; Patel, S.; Fletcher, A.; Ragan, I.C.; Leeson, P.D.; et al. 3-(1-Piperazinyl)-4,5-Dihydro-1H-Benzo[g]Indazoles: High Affinity Ligands for the Human Dopamine D4 Receptor with Improved Selectivity over Ion Channels. Bioorg. Med. Chem. 1998, 6, 743–753. [Google Scholar] [CrossRef]
  6. Reddy, S.T.; Shiva Kumar, K.; Meda, C.L.T.; Kandale, A.; Rambabu, D.; Rama Krishna, G.; Hariprasad, C.; Rao, V.; Venkataiah, S.; Malla Reddy, C.; et al. Conformationally Restricted Novel Pyrazole Derivatives: Synthesis of 1,8-Disubstituted 5,5-Dimethyl-4,5-Dihydro-1H-Benzo[g]Indazoles as a New Class of PDE4 Inhibitors. Bioorg. Med. Chem. Lett. 2012, 22, 3248–3255. [Google Scholar] [CrossRef] [PubMed]
  7. Deiana, V.; Gómez-Cañas, M.; Pazos, M.R.; Fernández-Ruiz, J.; Asproni, B.; Cichero, E.; Fossa, P.; Muñoz, E.; Deligia, F.; Murineddu, G.; et al. Tricyclic Pyrazoles. Part 8. Synthesis, Biological Evaluation and Modelling of Tricyclic Pyrazole Carboxamides as Potential CB2 receptor Ligands with Antagonist/Inverse Agonist Properties. Eur. J. Med. Chem. 2016, 112, 66–80. [Google Scholar] [CrossRef]
  8. Aparicio, D.A.; Lobo, G.M.; Sánchez, J.L.; Monasterios, M.C.; Acosta, M.E.; Lima, L.; Llovera, L.J.; Ángel, J.E.; Charris, J.E. Synthesis of Trans- and Cis-2-Acetyl-3-Phenyl-3,3a,4,5-Tetrahydro-2H-Benzo[g] Indazoles: Evaluation as Inhibitors of β-Hematin Formation. J. Chem. Res. 2017, 41, 668–672. [Google Scholar] [CrossRef]
  9. Jakob, C.G.; Upadhyay, A.K.; Donner, P.L.; Nicholl, E.; Addo, S.N.; Qiu, W.; Ling, C.; Gopalakrishnan, S.M.; Torrent, M.; Cepa, S.P.; et al. Novel Modes of Inhibition of Wild-Type Isocitrate Dehydrogenase 1 (IDH1): Direct Covalent Modification of His315. J. Med. Chem. 2018, 61, 6647–6657. [Google Scholar] [CrossRef]
  10. Gautam, P.; Gautam, D.; Chaudhary, R.P. Experimental and Theoretical Investigations on Acid Catalysed Stereoselective Synthesis of New Indazolyl-Thiazole Derivatives. J. Mol. Struct. 2018, 1160, 333–341. [Google Scholar] [CrossRef]
  11. Maurya, H.K.; Verma, R.; Alam, S.; Pandey, S.; Pathak, V.; Sharma, S.; Srivastava, K.K.; Negi, A.S.; Gupta, A. Studies on Substituted Benzo[h]Quinazolines, Benzo[g]Indazoles, Pyrazoles, 2,6-Diarylpyridines as Anti-Tubercular Agents. Bioorg. Med. Chem. Lett. 2013, 23, 5844–5849. [Google Scholar] [CrossRef] [PubMed]
  12. Gautam, D.; Chaudhary, R.P. Synthesis, Structure and Antimicrobial Evaluation of New 3,3a,4,5-Tetrahydro-2H-Benzo[g]Indazol-2-yl-Thiazol-4(5H)-Ones. Spectrochim. Acta.-Part. A Mol. Biomol. Spectrosc. 2015, 135, 219–226. [Google Scholar] [CrossRef] [PubMed]
  13. Tzanetou, E.; Liekens, S.; Kasiotis, K.M.; Fokialakis, N.; Haroutounian, S.A. Novel Pyrazole and Indazole Derivatives: Synthesis and Evaluation of Their Anti-Proliferative and Anti-Angiogenic Activities. Arch. Pharm. 2012, 345, 804–811. [Google Scholar] [CrossRef] [PubMed]
  14. Zong, X.; Cai, J.; Chen, J.; Sun, C.; Li, L.; Ji, M. Discovery of 3,3a,4,5-Tetrahydro-2H-Benzo[g]Indazole Containing Quinoxaline Derivatives as Novel EGFR/HER-2 Dual Inhibitors. RSC Adv. 2015, 5, 24814–24823. [Google Scholar] [CrossRef]
  15. Kasiotis, K.M.; Tzanetou, E.N.; Stagos, D.; Fokialakis, N.; Koutsotheodorou, E.; Kouretas, D.; Haroutounian, S.A. Novel Conformationally Constrained Pyrazole Derivatives as Potential Anti-Cancer Agents. Z. Naturforsch. B. 2015, 70, 677–690. [Google Scholar] [CrossRef]
  16. Laroum, R.; Berrée, F.; Roisnel, T.; Dorcet, V.; Carboni, B.; Debache, A. AlCl3-Promoted Reaction of Cycloalkanones with Hydrazones: A Convenient Direct Synthesis of 4,5,6,7-Tetrahydro-1H-Indazoles and Their Analogues. Z. Tetrahedron Lett. 2019, 60, 150988. [Google Scholar] [CrossRef]
  17. Dark, G.; Hill, S.A.; Prise, S.; Tozer, G.; Pettit, G.; Chaplin, D. Combretastatin A-4, an Agent That Displays Selective Toxicity towards Tumour Vasculature. Cancer Res. 1997, 33, 832–835. [Google Scholar] [CrossRef]
  18. Porcù, E.; Bortolozzi, R.; Basso, G.; Viola, G. Recent Advances in Vascular Disrupting Agents in Cancer Therapy. Futur. Med. Chem. 2014, 6, 1485–1498. [Google Scholar] [CrossRef]
  19. Pérez-Pérez, M.J.; Priego, E.M.; Bueno, O.; Martins, M.S.; Canela, M.D.; Liekens, S. Blocking Blood Flow to Solid Tumors by Destabilizing Tubulin: An Approach to Targeting Tumor Growth. J. Med. Chem. 2016, 59, 8685–8711. [Google Scholar] [CrossRef]
  20. Devkota, L.; Lin, C.M.; Strecker, T.E.; Wang, Y.; Tidmore, J.K.; Chen, Z.; Guddneppanavar, R.; Jelinek, C.J.; Lopez, R.; Liu, L.; et al. Design, Synthesis, and Biological Evaluation of Water-Soluble Amino Acid Prodrug Conjugates Derived from Combretastatin, Dihydronaphthalene, and Benzosuberene-Based Parent Vascular Disrupting Agents. Bioorg. Med. Chem. 2016, 24, 938–956. [Google Scholar] [CrossRef]
  21. Jiang, J.; Zhang, H.; Wang, C.; Zhang, Q.; Fang, S.; Zhou, R.; Hu, J.; Zhu, J.; Zhou, Y.; Luo, C.; et al. 1-Phenyl-Dihydrobenzoindazoles as Novel Colchicine Site Inhibitors: Structural Basis and Antitumor Efficacy. Eur. J. Med. Chem. 2019, 177, 448–456. [Google Scholar] [CrossRef] [PubMed]
  22. Bueno, O.; Tobajas, G.; Quesada, E.; Estévez-Gallego, J.; Noppen, S.; Camarasa, M.J.; Díaz, J.F.; Liekens, S.; Priego, E.M.; Pérez-Pérez, M.J. Conformational Mimetics of the α-Methyl Chalcone TUB091 Binding Tubulin: Design, Synthesis and Antiproliferative Activity. Eur. J. Med. Chem. 2018, 148, 337–348. [Google Scholar] [CrossRef] [PubMed]
  23. Ramírez-Prada, J.; Robledo, S.M.; Vélez, I.D.; Crespo, M.d.P.; Quiroga, J.; Abonia, R.; Montoya, A.; Svetaz, L.; Zacchino, S.; Insuasty, B. Synthesis of Novel Quinoline-based 4,5-dihydro-1H-pyrazoles as Potential Anticancer, Antifungal, Antibacterial and Antiprotozoal Agents. Eur. J. Med. Chem. 2017, 131, 237–254. [Google Scholar] [CrossRef] [PubMed]
  24. Romo, P.E.; Isaza, J.H.; Insuasty, B.; Abonia, R.; del Crespo, M.P.; Quiroga, J. Synthesis of Pyrazolo[3,4-b]Azepines and Their Antioxidant and Antibacterial Studies. Monatsh. Chem. 2019, 150, 1503–1511. [Google Scholar] [CrossRef]
  25. Crespo, M.P.; Avery, T.D.; Hanssen, E.; Fox, E.; Robinson, T.V.; Valente, P.; Taylor, D.K.; Tilley, L. Artemisinin and a Series of Novel Endoperoxide Antimalarials Exert Early Effects on Digestive Vacuole Morphology. Antimicrob. Agents Chemother. 2008, 52, 98–109. [Google Scholar] [CrossRef]
  26. Crespo, M.P.; Wei, M.Q. Antitumor Activity of Artemisinin and Its Derivatives: From a Well-Known Antimalarial Agent to a Potential Anticancer Drug. J. Biomed. Biotechnol. 2012, 2012, 93–102. [Google Scholar] [CrossRef]
  27. Meyer, M.D.; Altenbach, R.J.; Hancock, A.A.; Buckner, S.A.; Knepper, S.M.; Kerwin, J.F. Synthesis and in Vitro Characterization of N-[5-(4,5-Dihydro-1H-Imidazol-2-Yl)- 2-Hydroxy-5,6,7,8-Tetrahydronaphthalen-1-Yl]Methanesulfonamide and Its Enantiomers: A Novel Selective α1A Receptor Agonist. J. Med. Chem. 1996, 39, 4116–4119. [Google Scholar] [CrossRef]
  28. Jagtap, P.; Alexei, D.; Sungwoon, C.; Heather, K.; Junying, Y.; Cuny, G.D. Structure-Activity Relationship Study of Tricyclic Necroptosis Inhibitors. J. Med. Chem. 2007, 50, 1886–1895. [Google Scholar] [CrossRef]
  29. Stewart, J.J.P. MOPAC 2012; Stewart Computational Chemistry: Colorado Springs, CO, USA; Available online: http://OpenMOPCAC.net (accessed on 20 November 2019).
  30. Schmitt, D.M.; Connolly, K.L.; Jerse, A.E.; Detrick, M.S.; Horzempa, J. Antibacterial Activity of Resazurin-Based Compounds against Neisseria Gonorrhoeae in Vitro and in Vivo. Int. J. Antimicrob. Agents 2016, 48, 367–372. [Google Scholar] [CrossRef]
  31. Tanaka, M.; Furuya, R.; Kobayashi, I.; Kanesaka, I.; Ohno, A.; Katsuse, A.K. Antimicrobial Resistance and Molecular Characterisation of Neisseria Gonorrhoeae Isolates in Fukuoka, Japan, 1996–2016. J. Glob. Antimicrob. Resist. 2019, 17, 3–7. [Google Scholar] [CrossRef]
  32. Costa-Lourenço, A.P.R.; Barros dos Santos, K.T.; Moreira, B.M.; Fracalanzza, S.E.L.; Bonelli, R.R. Antimicrobial Resistance in Neisseria Gonorrhoeae: History, Molecular Mechanisms and Epidemiological Aspects of an Emerging Global Threat. Braz. J. Microbiol. 2017, 48, 617–628. [Google Scholar] [CrossRef] [PubMed]
  33. Leber, A.L. Antimicrobial Susceptibility Testing. In Clinical Microbiology Procedures Handbook, 4th ed.; American Society for Microbiology Press: Washington, DC, USA, 2016. [Google Scholar]
  34. Clinical and Laboratory Standards Institute (CLSI). Performance Standards for Antimicrobial Susceptibility Testing: Twenty-third Informational Supplement, Document M100–S26, 26th ed.; CLSI: Wayne, PA, USA, 2008; pp. 1–250. [Google Scholar]
  35. Liaras, K.; Geronikaki, A.; Glamočlija, J.; Ćirić, A.; Soković, M. Thiazole-Based Chalcones as Potent Antimicrobial Agents. Synthesis and Biological Evaluation. Bioorg. Med. Chem. 2011, 19, 3135–3140. [Google Scholar] [CrossRef] [PubMed]
  36. Gonococcal Isolate Surveillance Project. Available online: http://www.cdc.gov/std/gisp/ (accessed on 10 september 2019).
  37. Conceição, K.; Konno, K.; Richardson, M.; Antoniazzi, M.M.; Jared, C.; Daffre, S.; Camargo, A.C.; Pimenta, D. Isolation and biochemical characterization of peptides presenting antimicrobial activity from the skin of Phyllomedusa hypochondrialis. Peptides 2006, 27, 3092–3099. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are not available from the authors.
Figure 1. (A) Structure of CA-4 and recently reported conformationally restricted colchicine binding site inhibitors; (B) tricyclic pyrazole derivatives here addressed.
Figure 1. (A) Structure of CA-4 and recently reported conformationally restricted colchicine binding site inhibitors; (B) tricyclic pyrazole derivatives here addressed.
Molecules 24 04236 g001
Scheme 1. Synthesis of the substituted chalcones 710. Reagents and conditions: (a) aromatic aldehyde, NaOH, EtOH, rt., 7 h.
Scheme 1. Synthesis of the substituted chalcones 710. Reagents and conditions: (a) aromatic aldehyde, NaOH, EtOH, rt., 7 h.
Molecules 24 04236 sch001
Scheme 2. General synthesis of tricyclic derivatives 1118a,b. Reagents and conditions: (a) NH2NH2·H2O, AcOH, reflux, 2 h. (b) Fe powder, HCl, EtOH:H2O, reflux, 1 h.
Scheme 2. General synthesis of tricyclic derivatives 1118a,b. Reagents and conditions: (a) NH2NH2·H2O, AcOH, reflux, 2 h. (b) Fe powder, HCl, EtOH:H2O, reflux, 1 h.
Molecules 24 04236 sch002
Figure 2. Overlap of the 3D structures of isomers 18a (3R,3aS) (purple), 18b (3R,3aR) (orange) and compound 5 (R) (green).
Figure 2. Overlap of the 3D structures of isomers 18a (3R,3aS) (purple), 18b (3R,3aR) (orange) and compound 5 (R) (green).
Molecules 24 04236 g002
Figure 3. Compound 18a with a (3R,3aS) configuration is shown in purple while compound 18b with a (3R,3aR) configuration is shown in orange.
Figure 3. Compound 18a with a (3R,3aS) configuration is shown in purple while compound 18b with a (3R,3aR) configuration is shown in orange.
Molecules 24 04236 g003
Table 1. Antiproliferative activity of benzo[g]indazole derivatives expressed in IC50 a (μM).
Table 1. Antiproliferative activity of benzo[g]indazole derivatives expressed in IC50 a (μM).
Comp.Capan-1HCT-116NCI-H460DND-41HL-60K-562Z-138
11a40.8 ± 5.029.9 ± 9.010.8 ± 1.6>10030.9 ± 14.846.5 ± 0.2>100
11b27.0 ± 4.025.3 ± 18.911.8 ± 1.6>10038.0 ± 10.134.5 ± 5.970.4 ± 14.8
12a42.9 ± 1.626.9 ± 4.85.4 ± 0.5>10042.5 ± 0.564.3 ± 17.949.7 ± 25.2
12b17.9 ± 3.536.0 ± 6.814.9 ± 2.9>10038.9 ± 2.259.7 ± 4.845.1 ± 27.5
13a73.4 ± 26.636.5 ± 1.230.9 ± 10.051.0 ± 7.642.7 ± 3.973.1 ± 3.139.7 ± 6.9
13b32.3 ± 11.148.9 ± 6.845.0 ± 1.754.4 ± 10.242.9 ± 1.977.6 ± 9.142.3 ± 8.4
14a57.8 ± 2.482.0 ± 9.043.7 ± 4.7>10028.6 ± 35.7>100>100
14b42.9 ± 12.3>10044.6 ± 0.8>10026.8 ± 36.6>100>100
15a45.1 ± 20.643.5 ± 6.042.1 ± 7.538.6 ± 7.255.9 ± 20.851.0 ± 29.243.9 ± 28.1
15b53.5 ± 18.8>10054.1 ± 4.650.1 ± 25.044.8 ± 27.685.9 ± 7.150.1 ± 25.0
16a36.7 ± 14.445.0 ± 11.539.3 ± 8.530.3 ± 7.438.7 ± 7.544.9 ± 21.850.2 ± 0.9
16b42.6 ± 3.966.1 ± 31.333.7 ± 10.8837.7 ± 6.465.9 ± 20.275.7 ± 21.048.0 ± 26.0
17a56.4 ± 11.662.2 ± 10.947.3 ± 15.342.8 ± 28.649.0 ± 25.5>100>100
17b60.7 ± 1.780.6 ± 9.743.9 ± 9.666.1 ± 18.665.8 ± 17.1>100>100
18a30.3 ± 9.045.2 ± 2.636.6 ± 10.450.6 ± 8.552.5 ± 16.3>10089.9 ± 5.0
18b32.7 ± 22.052.1 ± 15.344.4 ± 2.240.8 ± 3.341.6 ± 5.069.2 ± 4.360.1 ± 1.8
Docetaxel0.0042 ± 0.00210.0009 ± 0.00080.0038 ± 0.00290.0033 ± 0.00140.0023 ± 0.00030.0037 ± 0.00030.0011 ± 0.0008
Stauroporine0.0007 ± 0.00020.00010 ± 0.00000.0015 ± 0.00040.0043 ± 0.00220.0074 ± 0.00170.0224 ± 0.00740.0003 ± 0.0001
Capan-1: pancreatic adenocarcinoma; Hap1: chronic myeloid leukemia; HCT-116: colorectal carcinoma; NCI-H460: lung carcinoma; DND-41: acute lymphoblastic leukemia; HL-60: acute myeloid leukemia; K-562: chronic myeloid leukemia; Z-138: non-Hodgkin lymphoma. a IC50: Concentration of each compound that inhibits 50% of cell proliferation. Mean value of two experiments ±SD.
Table 2. In vitro antibacterial activity of nitro derivatives expressed in MIC a (μg/mL).
Table 2. In vitro antibacterial activity of nitro derivatives expressed in MIC a (μg/mL).
Comp.S. aureus
ATCC 25923
S. aureus
ATCC 43300
VISAE. coli
ATCC 25922
P. aeruginosa
ATCC 27853
K. pneumoniae
ATCC700603
K. pneumoniae
BAA 1705
N. gonorrhoeae
ATCC 49226
7>1000>1000>1000>1000>1000>1000>1000≥500
8>1000>1000>1000>1000>1000>1000>1000>1000
9>1000>1000>1000>1000>1000>1000>1000>1000
10>1000>1000>1000>1000>1000>1000>1000>1000
11a>1000>1000>1000>1000>1000>1000>1000≥500
11b>1000>1000>1000>1000>1000>1000>1000≥500
12a>1000>1000>1000>1000>1000>1000>1000250
12b>1000>1000>1000>1000>1000>1000>1000>1000
13a>1000>1000>1000>1000>1000>1000>1000≥500
13b>1000>1000>1000>1000>1000>1000>100062.5
14a>1000>1000>1000>1000>1000>1000>1000≥500
14b>1000>1000>1000>1000>1000>1000>1000≥500
GEN b<1<0.25<0.25<0.62510.625<0.3121.25–2.5
TET c 1
a MIC: Minimum inhibitory concentration of each compound that inhibit visible bacterial growth. MIC values > 1000 mg/mL were considered as not relevant or not active. Antibacterial drug control: b GEN: Gentamicin and c TET: Tetracycline.

Share and Cite

MDPI and ACS Style

Cuartas, V.; Crespo, M.d.P.; Priego, E.-M.; Persoons, L.; Daelemans, D.; Camarasa, M.-J.; Insuasty, B.; Pérez-Pérez, M.-J. Design and Synthesis of New 6-Nitro and 6-Amino-3,3a,4,5-Tetrahydro-2H-Benzo[g]indazole Derivatives: Antiproliferative and Antibacterial Activity. Molecules 2019, 24, 4236. https://doi.org/10.3390/molecules24234236

AMA Style

Cuartas V, Crespo MdP, Priego E-M, Persoons L, Daelemans D, Camarasa M-J, Insuasty B, Pérez-Pérez M-J. Design and Synthesis of New 6-Nitro and 6-Amino-3,3a,4,5-Tetrahydro-2H-Benzo[g]indazole Derivatives: Antiproliferative and Antibacterial Activity. Molecules. 2019; 24(23):4236. https://doi.org/10.3390/molecules24234236

Chicago/Turabian Style

Cuartas, Viviana, María del Pilar Crespo, Eva-María Priego, Leentje Persoons, Dirk Daelemans, María-José Camarasa, Braulio Insuasty, and María-Jesús Pérez-Pérez. 2019. "Design and Synthesis of New 6-Nitro and 6-Amino-3,3a,4,5-Tetrahydro-2H-Benzo[g]indazole Derivatives: Antiproliferative and Antibacterial Activity" Molecules 24, no. 23: 4236. https://doi.org/10.3390/molecules24234236

Article Metrics

Back to TopTop