Next Article in Journal
Behavior of Longitudinal Plate-to-Rectangular Hollow Structural Section K-Connections Subjected to Cyclic Loading
Previous Article in Journal
Additive Manufacturing of Reinforced Concrete—Development of a 3D Printing Technology for Cementitious Composites with Metallic Reinforcement
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pd(PPh3)4 Catalyzed Synthesis of Indazole Derivatives as Potent Anticancer Drug

by
Jagan Mohana Rao Saketi
1,
S. N. Murthy Boddapati
1,
Raghuram M.
2,
Syed Farooq Adil
3,*,
Mohammed Rafi Shaik
3,
Osamah Alduhaish
3,*,
Mohammed Rafiq H. Siddiqui
3 and
Hari Babu Bollikolla
1,*
1
Department of Chemistry, Acharya Nagarjuna University, Guntur 522510, India
2
Department of Botany & Microbiology, Acharya Nagarjuna University, Guntur 522510, India
3
Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Appl. Sci. 2020, 10(11), 3792; https://doi.org/10.3390/app10113792
Submission received: 29 April 2020 / Revised: 25 May 2020 / Accepted: 26 May 2020 / Published: 29 May 2020
(This article belongs to the Section Chemical and Molecular Sciences)

Abstract

:
A series of 3-aryl indazoles and 1-methyl-3-aryl indazole derivatives are prepared with exceptional yields by coupling with several arylboronic acids and methylation by two dissimilar approaches. The as-prepared indazole derivatives (3a–3j) and their N-methyl derivatives (5a–5j) are evaluated for in vitro anticancer activity against two cancer cell lines, HCT-116 and MDA-MB-231. The results reveal that the indazole derivatives tested display mild to moderate anticancer activities against the cell lines tested.

1. Introduction

Cancer, a disease of the cell cycles, has remained the largest cause of mortality and morbidity for several decades now. In spite of rapid development in diagnostic and therapeutic protocols, according to the World Health Organization (WHO), cancer holds second place after cardiovascular disease as a cause of death in the world [1]. Among all the available therapeutic methods, chemotherapy still remains a significant option for the treatment of cancer, which has emerged as a new era of molecularly targeted therapeutics [2]. However, the major drawback of successful cancer treatment is the emergence of multi-drug resistance (MDR) in various cancer cell lines due to mutations in the cell, which limit the successful outcomes in most of the cases. Consequently, there is a requirement of novel advances that are exclusively designed to overcome the menace of drug resistance. Thus, the development of novel, potent, and selective anticancer agents is still one of the most significant areas of modern cancer research and turns out to be the main objective of organic and medicinal chemists across the world.
Among the various organic molecules, the nitrogen-containing heterocycles are important building blocks for many bioactive natural products and commercially available drugs. Indazole and its derivatives belong to an enormously important family of nitrogen-containing heterocyclic systems (Figure 1), often allied with a wide range of biological activities [3,4,5,6,7]. Many of indazole derivatives have been reportedly found to possess potent pharmacological activity such as anti-tumor [8,9], anti-platelet [10], antiviral [11], antioxidant [12], anti-spermatogenic activity [13], anti-tubercular [14,15], anti-inflammatory and anti-microbial [16], neuroprotection [17], and COX inhibition activity [18]. Moreover, some indazoles were reported as protein kinase C-B/AKt inhibitors [19], potent IDO1/TDO dual inhibitors [20], and also as 5-HT2, 5-HT3, and 5-HT4 receptor antagonists [21,22].
Furthermore, many of the synthetic and natural indazole based heterocycles with a sundry mechanism of action have been reported as lead anticancer agents [23,24,25]. Importantly, a number of indazole based anticancer drugs (Figure 2) were used clinically. For example, niraparib has been widely used as an anticancer drug for the treatment of recurrent epithelial ovarian, fallopian tube, breast, and prostate cancer [26], pazopanib [27] and axitinib [28] are tyrosine kinase inhibitors approved by the FDA for renal cell carcinoma; these are some of the present-day anticancer drugs possessing a privileged indazole skeleton. These interesting therapeutic properties of indazole derivatives have made them attractive target molecules in organic synthesis.
Considering this wide range of pharmacological activities of indazole scaffolds, various methodologies have been developed by the researchers for the synthesis of these moieties, of which many employed transition metal-based catalytic systems. The various metal catalysts that have been employed are [Ir(OMe)(COD)]2 [29], [Cp*RhCl2]2 [30,31], CuCl [32], [Cp*CoCl2]2 [33], Pd(OAc)2 [34], Pd(dba)2 [35], ZnBr [36], FeBr3 [37], and NiCl2 [38]. Very recently, our group developed efficient methodologies towards the construction of various heterocyclic compounds using Cu catalyst [39,40,41,42,43]. Inspired by the above facts, and in continuation of our efforts towards the development of new heterocyclic moieties of therapeutic importance [44,45,46], in the present investigation, we report the synthesis of a series of 3-aryl-1H- indazoles and N-methyl-3-arylindazoles using Pd(PPh3)4 catalyst as shown in Scheme 1. The as-synthesized compounds are characterized using spectroscopic techniques such as FT-IR, 1H NMR, 13C NMR, and ESI-MS and then later evaluated for their cell growth inhibitory activities (IC50) against HCT-116 and MDA-MB-231 cancer cell lines using the MTT assay method.
Reagents and conditions: (i) KOH, I2, DMF, 25 °C, 2 h, 77%, (ii) Ar‒B(OH)2, Pd(PPh3)4, NaHCO3, DMF, 80 °C, 8-12 h, 55-70%, (iii) MeI, KOH, acetone, 0 °C, 10–12 h, 58%–75% (iv) Ar‒B(OH)2, Pd(PPh3)4, NaHCO3, DMF, 80 °C, 8–12 h, 58%‒75%.

2. Experimental

General Information: All the chemicals and reagents are obtained from Sigma-Aldrich(Merck), Karnataka, India; S. D. Fine, Tamilnadu, India, Spectrochem, Mumbai, India, and are utilized without any further purification. Solvents used are dried prior to their use. Reactions are monitored by using precoated (Kieselgel 60 F254, Merck) TLC silica gel plates. Column chromatography is performed using silica gel (60–120 mesh, Merck). Cintex melting point apparatus is used to determine the Melting points. Perkin Elmer 400 FT-IR spectrometer (υmax in cm−1) or a Varian 670-IR FT-IR spectrometer (ATR) is used to record the IR (KBr) spectra. A Bruker DRX-300 (300 MHz FT NMR) or Varian Mercury 500 MHz spectrometer were utilized in recording the 1H NMR and 13C NMR spectra in CDCl3 and DMSO-d6. Chemical shifts are presented in δ ppm employing TMS as an internal reference. A Jeol SX-102 spectrometer is used to record the mass spectra.

2.1. Synthesis of 3-iodo-1H-indazole (2)

To a stirred solution of indazole (1) (0.2 g, 1.69 mmol) in DMF (10 mL), iodine (0.8 g, 3.38 mmol) was added, followed by addition of KOH pellets (0.3 g, 6.77 mmol) and the whole reaction mixture was stirred for 1 h at room temperature. Then, the mixture was poured into 10% aqueous NaHSO3 and extracted by diethyl ether. Organic layer was washed with water and saturated brine solution, dried over sodium sulphate and subjected to removal of solvent, which yielded a white solid 3-iodo-1H-indazole (2) (77%); m.p. 90–92 °C; IR (KBr, υmax, cm−1): 3311 (NH str), 3046 (ArCH str), 1650, 1596, 1558 (ArC=C str), 1415 (C=N str), 771 (C-I str); 1H NMR (500 MHz, CDCl3): δ 10.5 (s, 1H, NH), 7.52–7.43 (m, 3H, Ar-H), 7.25–7.21 (d, J = 10 Hz, 1H, Ar-H); 13C NMR (75 MHz, CDCl3): δ = 138.6, 132.8, 130.7, 128.9, 126.7, 122.0. HRMS: (ESI m/z) 244.9 (M + H)+.

2.2. General Procedure for Synthesis of 3-arylindazole (3a3j)

Under nitrogen atmosphere, to the mixture of 3-iodo indazole (2) (0.3 g, 1.2 mmol) and aryl boronic acid (1.8 mmol) in DMF (60 mL), NaHCO3 solution (0.3 g, 3.6 mmol) (2:1 DMF–water) was added. To this reaction mixture, Pd(PPh3)4 (0.14 g, 0.12 mmol) was added and refluxed at 80 °C for 8–12 h with vigorous stirring. The reaction mixture was then subjected to evaporation under vacuum to obtain a dry product, which was then dissolved in ethyl acetate and washed with saturated brine solution, dried over sodium sulfate, and the solvent ethyl acetate was removed under vacuum to give crude. The crude mixture was purified by silica gel (60–120 mesh) column chromatography using 20% ethyl acetate in hexane as eluent to afford the corresponding 3-aryl-1H-indazoles (3a3j).

2.3. General Procedure for Synthesis of N-methyl-3-aryl indazole (5a5j) (Route 1)

A solution of 3-aryl indazole (3) (0.66 mmol) in acetone was cooled to 0 °C, and to it was added KOH (0.05 g, 1.00 mmol). After 15 minutes, at 0 °C, methyl iodide (0.04 mL) is added and stirred for 2 h. The solvent from the reaction mixture was evaporated; the crude solid obtained was dissolved in ethyl acetate and washed with water and brine, dried over sodium sulfate and the solvent removed under vacuum to give a crude compound N-methyl-3-aryl indazole. The crude mixture was purified by silica gel (60–120 mesh) column chromatography using 20% ethylacetate in hexane as eluent to afford corresponding N-methyl-3-aryl indazole (5a5j).

2.4. General Procedure for Synthesis of N-methyl-3-aryl indazole (5a5j) (Route 2)

Under nitrogen atmosphere, to a mixture of N-methyl-3-iodoindazole 4 (0.3 g, 1.16 mmol) and aryl boronic acid (1.74 mmol) in DMF, NaHCO3 (0.2 g, 3.4 mmol) in water was added. To this reaction, mixture Pd (PPh3)4 (0.1 g, 0.1 mmol) was added and refluxed with vigorous stirring for 10–14 h. The solvent of the reaction mixture was then evaporated, and the crude obtained was dissolved in ethyl acetate; the organic phase was washed with brine solution, dried over sodium sulfate, and the solvent was removed under vacuum to give crude compound. The crude mixture is purified by silica gel (60–120 mesh) column chromatography using 30% ethyl acetate in hexane as eluent to afford the respective products N-methyl-3-aryl indazole (5a5j).

Spectral Data of Compounds (3a–3j) and (5a–5j)

3-Phenyl-1H-indazole(3a): m.p. 114–116 °C; IR (KBr, υmax, cm−1): 3691 (OH str), 3440 (b,NH str), 2991 (Ar=CH str), 2897 (CH str), 1601, 1545, 1498 (Ar C=C str), 1450 (C=N str), 1233 (N-N str); NMR: 1H (500 MHz, CDCl3): δ = 11.18 (b, 1H), 8.08–8.04 (m, 3H), 7.58 (t, J = 7.5, 2H), 7.49–7.46 (m, 1H), 7.43–7.37 (m, 2H), 7.29–7.25 (m, 1H); 13C-NMR: (125 MHz, DMSO): δ = 145.8, 141.7, 133.6, 128.9, 128.2, 127.7, 126.8, 121.4, 121.1, 121.0, 110.2; m/z (ESI-MS) 195.23 (M + H)+.
3-(Naphthalen-1-yl)-1H-indazole(3b): m.p. 136–138 °C; IR (KBr, υmax, cm−1): 3446 (b,NH str), 2929 (Ar=CH str), 1595, 1581, 1506 (ArC=C str), 1418 (C=N str), 1246 (N-N str);NMR: 1H (500 MHz, CDCl3): δ = 12.25 (b, 1H), 8.32–8.31 (m, 1H), 8.01–7.98 (m, 2H), 7.79–7.67 (m, 2H), 7.57–7.52 (m, 2H), 7.48–7.39 (m, 2H), 7.21 (t, J = 6.7, 1H), 6.93–6.92 (m, 1H). 13C-NMR (125 MHz, CDCl3): δ = 143.4, 134.1, 131.9, 131.3, 128.8, 128.2, 128.1, 126.7, 126.4, 126.3, 126.1, 125.4, 121.9, 121.3, 110.9: m/z (ESI-MS) 245.05 (M + H)+.
3-(4-Fluorophenyl)-1H-indazole(3c): m.p. 112–113 °C; IR (KBr, υmax, cm−1): 3406 (NH str), 3078 (ArH str), 2924 (ArH str), 1625, 1563 (ArC=C str), 1440 (C=N str), 1370 (C=N str), 814 (C-F str); 1H NMR (300 MHz, CDCl3): δ 8.05 (m, 3H, ArH), 7.45 (m, 2H, ArH), 7.25 (m, 1H, ArH); 7.21 (m, 2H, ArH); 13C NMR (75 MHz, CDCl3): δ = 151.0, 147.2, 138.5, 130.1, 128.8, 126.2, 124.4, 122.1, 121.7, 113.8; m/z (ESI-MS) 213.27 (M + H)+.
3-(Pyridin-4-yl)-1H-indazole(3d): m.p. 101–103 °C; IR (KBr, υmax, cm−1): 3430 (NH str), 3032 (ArCH str), 1584, 1549, 1491(ArC=C str), 1373 (C=N str), 1211 (N-N str); 1H NMR (300 MHz, CDCl3): δ 7.73–7.62 (m, 4H, Ar-H), 7.62 (d, J = 8 Hz, 2H, Ar-H); 7.45 (d, J = 9 Hz, 2H, Ar-H); 13C NMR (125 MHz, CDCl3): δ = 148.8, 143.6, 138.5, 130.1, 128.8, 126.2, 124.4, 122.1, 121.7, 112.3; m/z (ESI-MS) 196.26 (M + H)+.
3-(Pyridin-3-yl)-1H-indazole(3e): m.p. 184–186 °C; IR (KBr, υmax, cm−1): 3409 (b, NH str), 3066 (ArCH str), 1589, 1560, 1512(ArC=C str), 1340 (C=N str), 1216 (N-N str); 1H NMR (300 MHz, CDCl3): δ 10.92 (s, 1H, NH), 9.25 (s, 1H, ArH), 8.65 (d, 1H, J = 7.5, Ar-H), 8.42 (d, 1H, J = 8.4 Hz, ArH); 8.05 (d, J = 7.5 Hz, 1H, Ar-H) 7.22–7.64 (m, 4H, ArH); 13C NMR (125 MHz, CDCl3): δ = 148.5, 139.6, 139.2, 128.8, 127.5, 126.2, 125.2, 124.4, 124.3, 123.0, 122.6, 122.2; m/z (ESI-MS) 196.26 (M + H)+.
3-(4-Methoxyphenyl)-1H-indazole(3f): m.p. 85–87 °C; IR (KBr, υmax, cm−1): 3429 (NH str), 3054 (ArH str), 2927 (CH str), 1583, 1487 (ArC=C str), 1438 (C=N str), 1375 (C=N str), 1169 (C-O-C str); 1H NMR (300 MHz, CDCl3): δ 8.26 (s, 1H, NH), 8.03 (d, 1H, J = 8.0, ArH), 7.85 (dd, 2H, J = 10.5, ArH), 7.46 (m, 2H, ArH); 7.22 (m, 1H, ArH), 7.03 (dd, 2H, J = 10.4, ArH); 13C NMR (75 MHz, CDCl3): δ = 159.6, 145.7, 141.8, 128.9, 126.6, 126.2, 121.1, 121.1, 114.4, 110.3, 55.3; m/z (ESI-MS) 225.31 (M + H)+.
3-(4-(Methylthio)phenyl)-1H-indazole(3g): m.p. 123-125 °C; IR (KBr, υmax, cm−1): 3378 (NH str), 3052 (ArH str), 2925 (CH str), 1600, 1521 (ArC=C str), 1346 (C=N str), 1106 (C-S-C str); 1H NMR (300 MHz, CDCl3): δ 8.16 (s, 1H, NH), 7.92 (d, 2H, J = 7.5, ArH), 7.52 (d, 1H, J = 8.4, ArH), 7.38-7.7.51 (m, 4H, ArH); 7.23 (s, 1H, ArH), 2.45 (s, 3H, CH3); 13C NMR (125 MHz, CDCl3): δ = 160.0, 145.7, 141.8, 134.7, 129.9, 126.8, 121.4, 121.0, 120.2, 114.2, 112.9, 110.3, 55.3.; m/z (ESI-MS) 241.30 (M + H)+.
3-(2-Methoxyphenyl)-1H-indazole(3h): m.p. 115–116 °C; IR (KBr, υmax, cm−1): 3325 (b, NH str), 3025 (ArCH str), 2921 (CH str), 1659, 1513, 1437(ArC=C str), 1370 (C=N str), 1212 (N-N str), 1148 (C-O-C str); 1H NMR (300 MHz, CDCl3): δ 7.81 (s, 1H, NH), 7.72 (d, 1H, J = 7.5 Hz, ArH), 7.52 (d, J = 8.5 Hz, 1H, Ar-H) 7.42 (m, 2H, ArH); 7.21 (m, 1H, ArH), 7.09 (m, 1H, ArH), 7.02 (d, 1H, J = 10.2 Hz, ArH), 3.8 (s, 3H, OCH3).13C NMR (125 MHz, CDCl3): δ = 157.3, 143.0, 141.4, 131.4, 129.7, 126.3, 122.2, 122.0, 120.9, 120.5, 111.4, 110.4, 55.4; m/z (ESI-MS) 225.30 (M + H)+.
4-(1H-indazol-3-yl)phenol(3i): m.p. 121–123 °C; IR (KBr, υmax, cm−1): 3414 (OH str), 3315 (b,NH str), 2925 (Ar=CH str), 1651, 1560, 1505(ArC=C str), 1414 (C=N str), 1219 (N-N str), 1093 (C-O str); 1H NMR (300 MHz, CDCl3): δ 13.12 (s, 1H, PhOH), 9.62 (s, 1H, NH), 8.01 (d, 1H, J = 8.4 Hz, ArH), 7.82 (d, J = 7.5 Hz, 2H, Ar-H), 7.49 (d, J = 10.2, 1H, ArH), 7.39 (d, J = 10.4, 1H, ArH), 7.22 (d, J = 8.0 Hz, 1H, ArH), 6.96 (m, 2H, ArH); 13C NMR (75 MHz, CDCl3): δ = 158.4, 151.4, 148.6, 138.8, 132.5, 134.8, 128.7, 125.9, 122.1, 113.5, 104.1; m/z (ESI-MS) 211.30 (M + H)+.
4-(1H-indazol-3-yl)-N,N-dimethylbenzamide(3j): m.p. 116–117 °C; IR (KBr, υmax, cm−1): 3456 (b, NH str), 2933 (Ar=CH str), 2835 (CH str), 1817 (CO str), 1649, 1530, 1488(ArC=C str), 1386(C=N str), 1210 (N-N str); 1H NMR (500 MHz, CDCl3): δ 9.86 (s, 1H, NH), 8.09 (m, 3H, ArH), 7.61 (m, 3H, Ar-H), 7.45 (d, J = 10.5 Hz, 1H, ArH); 7.28 (s, 1H, ArH), 3.12 (s, 6H, CH3); 13C NMR (100 MHz, CDCl3): δ = 166.3, 145.7, 135.2, 132.2, 130.4, 128.7, 128.6, 128.1, 127.9, 127.3, 126.5, 114.1, 36.4; m/z (ESI-MS) 266.35 (M + H)+.
1-Methyl-3-phenyl-1H-indazole(5a): m.p. 76–78 °C; IR (KBr, υmax, cm−1): 3429 (NH str), 3054 (ArH str), 2927 (CH str), 1582, 1487 (ArC=C str), 1438 (C=N str), 1375 (C=N str), 1169 (C-O-C str); 1H NMR (400 MHz, DMSO-d6) δ 4.12 (3H, s) 7.24 (1H, t, J = 7.6 Hz) 7.38–7.43 (1H, m) 7.46 (1H, t, J = 7.6 Hz) 7.52 (2H, t, J = 7.6 Hz) 7.70 (1H, d, J = 8.3 Hz) 7.98 (2H, d, J = 7.3 Hz) 8.07 (1H, d, J = 8.3 Hz); 13C NMR (100 MHz, DMSO-d6): δ = 142.4, 141.7, 133.8, 129.4, 128.2, 127.2, 126.7, 121.7, 121.3, 121.1, 110.6, 36.0; m/z (ESI-MS) 209.27 (M + H)+.
1-Methyl-3-(naphthalen-1-yl)-1H-indazole(5b): m.p. 137–139 °C; IR (KBr, υmax, cm−1): 3130 (w, Ar=CH str), 2955, 2922, 2863 (CH str), 1625, 1583, 1495(ArC=C str), 1384 (C=N str), 1234 (N-N str), 1157 (C-S-C str); 1H NMR (400 MHz, DMSO-d6) δ 4.20 (3H, s) 7.20 (1H, t, J = 7.5 Hz) 7.46–7.69 (5H, m) 7.76 (2H, d, J = 7.7 Hz) 8.04 (2H, d, J = 8.1 Hz) 8.29 (1H, d, J = 8.3 Hz); 13C NMR (100 MHz, DMSO-d6): δ = 142.5, 141.1, 134.2, 131.6, 130.5, 128.8, 128.3, 126.9, 126.8, 126.6, 126.3, 126.1, 123.1, 121.5, 121.0, 110.6, 36.1; m/z (ESI-MS) 259.33 (M + H)+.
3-(4-Fluorophenyl)-1-methyl-1H-indazole(5c): m.p. 98–100 °C; IR (KBr, υmax, cm−1): 3098 (Ar=CH str), 2935, 2888 (CH str), 1601, 1524, 1480 (ArC=C str), 1354 (C=N str), 1229 (N-N str), 825 (C-F str); 1H NMR (300 MHz, CDCl3): δ 7.92 (m, 3H, ArH), 7.42 (s, 2H, ArH), 7.21 (m, 3H, ArH), 4.13 (s, 3H, CH3); 13C NMR (100 MHz, CDCl3): δ = 162.8, 145.2, 139.1, 136.7, 132.2, 128.9, 128.7, 128.2, 123.4, 119.5, 117.6, 110.5, 35.2; m/z (ESI-MS) 227.31 (M + H)+.
1-Methyl-3-(pyridin-4-yl)-1H-indazole(5d): m.p. 101–103 °C; IR (KBr, υmax,cm−1): 3052(Ar=CH str), 2925 (CH str), 1612, 1508, 1459(ArC=C str), 1390 (C=N str), 1205(N-N str); 1H NMR (300 MHz, CDCl3): δ 8.71 (d, 2H, J = 8.5 Hz, ArH), 8.06 (d, 1H, J = 8.0 Hz, ArH), 7.92 (d, 2H, J = 7.5 Hz, Ar-H), 7.42 (d, J = 7.0 Hz, 2H, ArH); 7.26 (m, 1H, ArH), 4.19 (s, 3H, CH3); 13C NMR (75 MHz, CDCl3): δ = 166.1, 160.7, 145.7, 135.2, 132.2, 128.7, 128.6, 128.1, 127.9, 126.5, 36.4; m/z (ESI-MS) 210.41 (M + H)+.
1-Methyl-3-(pyridin-3-yl)-1H-indazole(5e): m.p. 74–76 °C; IR (KBr, υmax, cm−1): 3136 (Ar=CH str), 2949, 2873 (CH str), 1593, 1534(ArC=C str), 1348 (C=N str), 1189 (N-N str); 1H NMR (300 MHz, CDCl3): δ 9.22 (s, 1H, ArH), 8.62 (s, 1H, ArH), 8.36 (dd, 1H, J = 7.5 Hz, ArH), 8.02 (d, J = 7.0 Hz, 1H, ArH); 7.51 (m, 3H, ArH), 7.32 (s, 1H, ArH). 4.18 (s, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ = 153.6, 149.8, 141.9, 141.8, 137.3, 126.9, 123.7, 122.9, 122.0, 120.5, 110.4, 36.2; m/z (ESI-MS) 210.29 (M + H)+.
3-(4-Methoxyphenyl)-1-methyl-1H-indazole(5f): m.p. 112–114 °C;IR (KBr, υmax, cm−1): 3098 (Ar=CH str), 2960, 2921, 2872 (CH str), 1607, 1569, 1530(ArC=C str), 1350 (C=N str), 1201 (N-N str), 1164 (C-O-C str); 1H NMR (300 MHz, CDCl3): δ 8.02 (d, J = 8.0 Hz, 1H, ArH), 7.90 (d, 2H, J = 8.5 Hz, ArH), 7.42 (m, 2H, ArH), 7.23 (m, 1H, ArH); 7.08 (d, J = 8.5, 2H, ArH), 4.15 (s, 3H, OCH3), 3.91 (s, 3H, NCH3); 13C NMR (100 MHz, CDCl3): 13C- NMR (100 MHz, DMSO-d6) δ = 159.4, 142.4, 141.6, 128.4, 126.6, 126.4, 121.4, 121.3, 121.0, 114.8, 110.5, 55.7, 35.9; m/z (ESI-MS) 239.09 (M + H)+.
1-Methyl-3-(4-(methylthio)phenyl)-1H-indazole(5g): m.p. 99–101 °C; IR (KBr, υmax, cm−1): 3095 (Ar=CH str), 2956, 2922, 2864 (CH str), 1625, 1582, 1495 (ArC=C str), 1384 (C=N str), 1234 (N-N str), 1157 (C-S-C str); 1H NMR (300 MHz, CDCl3): δ 8.01 (d, J = 7.5 Hz, 1H, ArH), 7.89 (d, 2H, J = 8.0 Hz, ArH), 7.41 (m, 4H, ArH), 7.20 (m, 1H, ArH); 4.14 (s, 3H, NCH3), 2.58 (s, 3H, NCH3); 13C NMR (100 MHz, CDCl3): δ = 166.4, 160. 6, 145.7, 135.2, 132.2, 131.7, 128.6, 128.1, 127.9, 126.5, 119.7, 55.4, 36.3; m/z (ESI-MS) 255.32 (M + H)+.
3-(2-Methoxyphenyl)-1-methyl-1H-indazole(5h): m.p. 105–107 °C; IR (KBr, υmax, cm−1): 3053.97b (Ar=CH str), 2955, 2894 (CH str), 1614, 1520, 1459(ArC=C str), 1372 (C=N str), 1278 (N-N str), 1172 (C-O-C str); 1H NMR (300 MHz, CDCl3): δ 8.01 (d, J = 9.0 Hz, 1H, ArH), 7.78 (d, 1H, J = 8.5 Hz, ArH), 7.41 (m, 3H, ArH), 7.05-7.18 (m, 3H, ArH); 4.16 (s, 3H, OCH3), 3.82 (s, 3H, NCH3); 13C NMR (100 MHz, CDCl3): δ = 161.7, 143.6, 141.7, 137.6, 132.9, 132.2, 128.9, 128.1, 128.6, 123.3, 120.4, 116.0, 110.5, 55.2, 35.2; m/z (ESI-MS) 239.33 (M + H)+.
4-(1-Methylindazol-3-yl)phenol(5i): m.p. 241–243 °C; IR (KBr, υmax, cm−1): 3691(OH str), 3439 (NH str), 2991 (Ar=CH str), 2897 (CH str), 1601, 1545, 1498 (ArC=C str), 1449 (C=N str), 1233 (N-N str); 1H NMR (400 MHz, DMSO-d6) δ 4.06 (3H, s) 6.93 (2H, d, J = 8.6 Hz) 7.18 (1H, t, J = 7.5 Hz) 7.42 (1H, t, J = 7.6 Hz) 7.62 (1H, d, J = 8.6 Hz) 7.80 (2H, d, J = 8.6 Hz) 8.00 (1H, d, J = 8.2 Hz) 9.65 (1H, s); 13C NMR (100 MHz, DMSO-d6) δ = 157.7, 142.8, 141.6, 128.5, 126.5, 124.8, 121.4, 121.2, 120.9, 116.2, 110.3, 35.8; m/z (ESI-MS) 225.10 (M + H)+.
N,N-Dimethyl-4-(1-methyl-1H-indazol-3-yl)benzamide(5j): m.p. 110–112 °C; IR (KBr, υmax, cm−1): 3054 (Ar=CH str), 2234 (C-NO2 str), 1679 (CO str), 1583, 1487(ArC=C str), 1375 (C=N str), 1212 (N-N str); 1H NMR (300 MHz, CDCl3): δ 8.02 (m, 1H, ArH), 7.57 (d, 2H, J = 8.0 Hz, ArH), 7.43 (d, 3H, J = 8.4 Hz, ArH), 7.23 (s, 1H, ArH); 4.17 (s, 3H, NCH3), 3.12 (s, 6H, N(CH3)2; 13C NMR (100 MHz, CDCl3): δ 169.2, 149.6, 147.9, 130.4, 128.7, 127.5, 126.2, 125.6, 124.1, 121.9, 111. 3, 37.2, 36.3; m/z (ESI-MS) 280.35 (M + H)+.

2.5. Procedure for Anti-Cancer Activity

The MTT cell proliferation assay method was used to analyze the cell growth on a protocol of 48 h [47]. Human colorectal cancer cell lines (HCT-116 and MDA-MB-231) were procured from the National Centre for Cell Sciences (NCCS), Pune, India, and maintained in DMEM. The cell lines were cultured with DMEM supplemented with 10% FBS, L-glutamine, NaHCO3, and an antibiotic solution containing penicillin (100 U/mL) and streptomycin (100 μg/mL). The exponentially growing cells were seeded at 5 × 103 cells per well into 96-well plates. The culture medium was removed after 24 h incubation at 37 °C and restored with fresh medium containing the candidate compounds in different concentrations. Next, the cells were incubated for another 72 h. Then, 20 mL of MTT (3-(4,5- dimethyl thiazol-2-yl)-2,5-diphenyltetrazolium bromide) solution (5 mg/mL) was added to all wells and incubated for 4 h at 37 °C. The medium containing MTT was discarded, 150 mL of dimethyl sulfoxide (DMSO) was added to each well and the plates agitated until the dark blue crystals (formazan) had completely dissolved; the absorbance was measured using a microplate reader at a wavelength of 570 nm. Each concentration was analyzed in triplicate, and the experiment is repeated three times. The average 50% inhibitory concentration (IC50) is determined from the concentration-response curves according to the inhibition ratio for each concentration.

3. Results and Discussions

3.1. Chemistry

3-substituted indazoles are common components in a variety of biologically potent molecules possessing a pharmaceutical interest in a variety of therapeutic areas [10,48,49,50]. Hence, the functionalization of indazoles at the C-3 position is of immense interest. With the increasing applications of 3-aryl indazoles in the pharmaceutical industry and inspired by the literature of Suzuki couplings, in the present work, we append the aromatic moieties after iodination at C-3 position of indazole, followed by palladium-catalyzed C-C bond formation to obtain 3-aryl-1H-indazoles (3a–3j). However, the various N-methyl-3-aryl-indazoles derivatives (5a–5j) are obtained by the N-methylation reactions, as given in Scheme 1.
The 3-iodo indazole (2) is the key intermediate in this process, which is obtained by the iodination of indazole (1) using KOH/I2 in DMF. Most of the synthesized compounds originated with the Pd-catalyzed aryl coupling reaction of the 3-iodoindazole(2) [51] with diverse aromatic boronic acids in dimethylformamide (DMF) which yields 3-aryl-1H-indazoles (3a–3j). Moreover, the synthesis of N-methyl-3-aryl-indazoles derivatives (5a–5j) is attempted by two routes (Scheme 1). In route 1, the Pd promoted cross-coupling reaction of the 3-iodoindazole [52], with a variety of arylboronic acids under conventional activation producing the consecutive 3-aryl-1H-indazoles (3a–3j), and yield obtained was 55%–70%, which on methylation with methyl iodide gave the final desired N-methyl-3-aryl-indazoles derivatives (5a–5j) in 58%–75% yield. However, in route 2, the 3-iodo-indazole (2) intermediate is first subjected to N- methylation using MeI to yield N-methyl-3-iodo-indazole intermediate (4), which is then reacted with a variety of arylboronic acids to yield N-methyl-3-substituted indazoles (5a–5j) in good yields. The individual synthetic results such as reaction time, yield, and melting point for the compounds 3a–3j and 5a–5j were indicated in Table 1 and Table 2, respectively. In the synthetic course, during the methylation of 3-substituted indazoles using methyl iodide, yields the N-1 methylated product predominantly, and we did not observe any N-2 methyl isomer formation in the reaction mixture. It might be due to the reason that the direct methylation of indazoles in the presence of a base generally provides thermodynamically stable N-1 methylated products predominantly [53].
All the spectroscopic and analytical data of the synthesized compounds are in full agreement with the anticipated structures. For example, for the sample 5f, the appearance of the characteristic peak at 2960 (s), 1164 (s) cm−1 is owed to the existence of CH3, C-O-C groups. The IR stretching bands at 1350 (s) cm−1 and 1201 (m) cm−1 are because of C=N and due to N-N stretchings, peaks at 1607 (m), 1569 (s) and 1530 (w) cm−1 are due to the aromatic Ar-C=C stretching, and peak at 3098 (w) cm−1 are because of aromatic =CH stretching which is further confirmed by other spectral analysis. The 1H-NMR spectrum of 5f displayed signals at chemical shift values δ 3.91 (s, 3H),δ 4.15 (s, 3H), which are assigned to the N-CH3, O-CH3 groups, doublet at δ 8.02, 7.90 ppm, multiplet at δ 7.42, 7.23 ppm and another doublet at δ 7.08 ppm assigned for aromatic protons. This spectral data provides strong evidence to assign the structure of the compound (5f) as 3-(4-methoxyphenyl)-1-methyl-1H-indazole, which is further authenticated from its 13C-NMR spectrum, which reveals the existence of 13 different carbons in the compound. The peaks at δ 35.9 and δ 55.7 ppm are allocated to the N-CH3 and O-CH3 carbons, respectively, while the signal at δ 154.9 ppm is attributed to the C3 carbon of indazole core nuclei. The signals at δ 143-110 ppm are due to the presence of aromatic moiety in the molecule 5f, i.e., 3-(4-methoxyphenyl)-1-methyl-1H-indazole. The molecular ion peak at 239.09 (M + H)+ in its mass spectra (ESI) further supports the formation of compound 5f.

3.2. Cytotoxic Study

Initially, the prepared compounds 3a–3j are screened for their in-vitro anticancer activity against the human colon carcinoma cell line (HCT-116) and the human breast cancer cell line (MDA-MB-231), according to the literature protocol [47]. The cell lines are cultured with DMEM supplemented with 10% FBS, L-glutamine, sodium bicarbonate, and an antibiotic solution containing penicillin (100 U/mL) and streptomycin (100 μg/mL). All cell lines are maintained in culture at 37 °C in an atmosphere of 5% carbon dioxide. The as-synthesized indazoles 3a–3j are screened for in vitro cytotoxic activity against HCT-116 and MBA-MB-231 cell lines. The anticancer properties of these analogs, i.e., 3a–3j, are compared with the standard doxorubicin. IC50 values of the test compound for 24 h on each cell line are calculated and presented in Table 3.
It is evident from the results that the tested indazole compounds 3c, 3g, and 3i are found to be more potent against the HCT-116 cell line than the MDA-MB-231 cell line. However, the test compound 3j possesses potent cytotoxic activity against both the cell lines tested, i.e., HCT-116 and MDA-MB-231 at IC50 < 87 μg/mL. Very few of the 3-aryl-1-H-indazole compounds (3h and 3c) are found to be moderately active against the two cell lines tested. The test compound 3j showed good activity against the HCT-116 cell line. The test compounds 3c and 3h exhibited almost similar activity against the HCT-116 cell line with IC50 < 94 μg/mL. The compounds 3h and 3c are found to be moderately active against the cell line MDA-MB-231 with IC50 < 96 and IC50 < 103 μg/mL, respectively. Unfortunately, the compounds 3a, 3b, and 3e are found to be inactive against the tested two cell lines.
After the screening of the cytotoxic properties of 3-aryl indazoles 3a–3j, the studies were extended to the evaluation of cytotoxic properties of the final compound i.e., N-methyl-3-aryl indazoles 5a–5j against the cancer cell lines HCT-119 and MDA-MB-231, according to the same literature protocol [47] employed above. The IC50 values of the test compounds are compared with the standard doxorubicin and the results of investigation were presented in Table 4. The IC50 value of the standard doxorubicin is 1.2 μg/mL against the HCT-116 cell line, 0.3 μg/mL against the MDA-MB-231 cell line. The tested compounds 5a–5j showed their IC50 values in between 54.1–172.4 μg/mL. All the obtained compounds displayed moderate to mild cytotoxic than the standard as evident from their higher IC50 values. From the above studies, it can be found that doxorubicin is more potent than the tested compounds; however, the structure related activity study of tested compounds can be used to guide us to develop potent molecules. A graphical illustration for the obtained IC50 values for the compounds 3a–3j and 5a–5j is given in Figure 3.
The result of the present investigation reveals that few of the tested compounds have shown significant decrease in cell viability in two test cell lines. It is evident from the results that the N-methyl-3-aryl indazoles (5a–5j) are found to be more potent than 3a–3j against HCT-116 and MDA-MB-231 cell lines. The compounds 5c, 5d, 5f, 5g, and 5i exhibit significant cytotoxic activities against the two cell lines tested. However, the test compound 5c is found to be more potent against the cell lines HCT116 and MDA-MB-231 with IC50 < 64 and IC50 < 59 μg/mL, respectively. The test compounds 5d and 5i show potent activity against the cell line HCT-116 with IC50 < 63 μg/mL. The compounds 5g, 5f display moderate activity against the HCT-116 cell line with IC50 < 73 μg/mL. The differential activity among the cell lines may be due to the structure–activity relationship of the molecules. Exponentially growing cells were treated with different concentrations of indazole compounds for 24 h and cell growth inhibition is analyzed through MTT assay.

Structure Activity Relationship (SAR)

The cautious investigation of the relation between structures and anticancer activities data of the test compounds reveals the following assumption about SAR: (i) N-methyl-3-aryl indazoles 5a–5j show higher activity than 3-aryl substituted indazoles derivatives 3a–3j against the tested cell lines HCT-16 and MDA-MB-231; (ii) additional, slight enhancement of the cytotoxic activity of compounds 5a–5j over 3a–3j can be attributed to the presence of the methyl group; (iii) the presence of electron withdrawing fluoro substitution at 4th position of the phenyl ring 5c is responsible for its superior anti cancer activity. In addition, electron releasing hydroxy group 5i and N,N-dimethylamide group 5j at para position of the phenyl ring exhibited good anti-cancer activity. Hence, from the results of above anticancer activity, it can be concluded that diverse structural requirements are essential for a compound to be active against different cancer targets.

4. Conclusions

In conclusion, a series of 3-aryl-1H-indazoles and N-methyl-3H-indazoles were synthesized successfully using simple reagents. All the synthesized indazoles were screened for their in vitro anti-cancer activities against the cell lines HCT-116 and MDA-MB-231. The results of the cytotoxic studies of the tested compounds reveal that compound 5c exhibited significant inhibitory effect on the two tested cancer cell lines amongst all the compounds synthesized. Compounds 5i, 5d, and 3j also exhibited good cytotoxic activity. Most of the compounds are active against human colon carcinoma cell line (HCT-116) and human breast cancer cell line (MDA-MB-231). However, the prepared compounds are comparatively less potent than commercially available drug doxorubicin (positive control). Nevertheless, we believe that slight structural modification of these active derivatives may yield better prospective anticancer drugs and demand further experimental investigations, especially in the area of anticancer research.

Supplementary Materials

Supplementary File 1

Author Contributions

Conceptualization, H.B.B.; methodology, J.M.R.S.; investigation, S.N.M.B.; carried out the preparation and characterization of Indazole derivatives, S.N.M.B. and R.M.; carried out the interpretation of some part of results, S.F.A., O.A., M.R.S. and H.B.B.; writing—original draft preparation, S.N.M.B.; J.M.R.S.; writing—review and editing, H.B.B.; helped to draft the manuscript, J.M.R.S., S.F.A., O.A., M.R.S. and H.B.B.; supervision, H.B.B. and M.R.H.S.; All authors have read and agreed to the published version of the manuscript.

Acknowledgments

The authors extend their appreciation to the Deanship of Scientific Research at King Saud University for funding this work through the research group project No. RG-1440-068.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tantawy, M.A.; Nafie, M.S.; Elmegeed, G.A.; Ali, I.A.I. Auspicious role of the steroidal heterocyclic derivatives as a platform for anti-cancer drugs. Bioorg. Chem. 2017, 73, 128–146. [Google Scholar]
  2. MacDonald, V. Chemotherapy: Managing side effects and safe handling. Can. Vet. J. 2009, 50, 665–668. [Google Scholar]
  3. Shimada, I.; Maeno, K.; Kazuta, K.-I.; Kubota, H.; Kimizuka, T.; Kimura, Y.; Hatanaka, K.-I.; Naitou, Y.; Wanibuchi, F.; Sakamoto, S. Synthesis and structure–activity relationships of a series of substituted 2-(1H-furo [2,3-g] indazol-1-yl) ethylamine derivatives as 5-HT2C receptor agonists. Bioorg. Med. Chem. 2008, 16, 1966–1982. [Google Scholar]
  4. Li, X.; Chu, S.; Feher, V.A.; Khalili, M.; Nie, Z.; Margosiak, S.; Nikulin, V.; Levin, J.; Sprankle, K.G.; Tedder, M.E.; et al. Structure-Based Design, Synthesis, and Antimicrobial Activity of Indazole-Derived SAH/MTA Nucleosidase Inhibitors. J. Med. Chem. 2003, 46, 5663–5673. [Google Scholar] [CrossRef]
  5. 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]
  6. Di Cosimo, S.; Ferretti, G.; Papaldo, P.; Carlini, P.; Fabi, A.; Cognetti, F. Lonidamine: Efficacy and safety in clinical trials for the treatment of solid tumors. Drugs Today (Barc) 2003, 39, 157–174. [Google Scholar]
  7. Cerecetto, H.; Gerpe, A.; González, M.; Aran, V.J.; de Ocariz, C.O. Pharmacological properties of indazole derivatives: Recent developments. Mini-Rev. Med. Chem. 2005, 5, 869–878. [Google Scholar] [CrossRef]
  8. Srinivas, A.; SriRamya, P.V.; Angeli, A.; Supuran, C.T.; Arifuddin, M. Novel sulfocoumarin/coumarin/4-sulfamoylphenyl bearing indazole-3-carboxamide hybrids: Synthesis and Selective inhibition of tumor associated carbonic anhydrase isozymes IX and XII. Chem. Med. Chem 2017, 12, 1578–1584. [Google Scholar]
  9. Shin, D.H.; Kim, J.H.; Jung, Y.J.; Kim, K.E.; Jeong, J.M.; Chun, Y.S.; Park, J.W. Preclinical evaluation of YC-1, a HIF inhibitor, for the prevention of tumor spreading. Cancer Lett. 2007, 255, 107–116. [Google Scholar] [CrossRef]
  10. Lee, F.Y.; Lien, J.C.; Huang, L.J.; Huang, T.M.; Tsai, S.C.; Teng, C.M.; Wu, C.C.; Cheng, F.C.; Kuo, S.C. Synthesis of 1-benzyl-3-(5′-hydroxymethyl-2′-furyl) indazole Analogues as Novel Antiplatelet Agents. J. Med. Chem. 2001, 44, 3746–3749. [Google Scholar] [CrossRef]
  11. Shi, J.J.; Ji, F.H.; He, P.L.; Yang, Y.X.; Tang, W.; Zuo, J.P.; Li, Y.C. Synthesis and Hepatitis C Antiviral Activity of 1-Aminobenzyl-1H-indazole-3-carboxamide Analogues. ChemMedChem 2013, 8, 722–725. [Google Scholar] [CrossRef]
  12. Sapnakumari, M.; Narayana, B.; Sarojini, B.K.; Madhu, L.N. Synthesis of new indazole derivatives as potential antioxidant agents. Med. Chem. Res. 2014, 23, 2368–2376. [Google Scholar] [CrossRef]
  13. Corsi, G.; Palazzo, G.; Germani, C.; Scorza Barcellona, P.; Silvestrini, B. 1-Halobenzyl-1H-indazole-3-carboxylic acids. A new class of antispermatogenic agents. J. Med. Chem. 1976, 19, 778–783. [Google Scholar] [CrossRef]
  14. Karalı, N.; Gürsoy, A.; Kandemirli, F.; Shvets, N.; Kaynak, F.B.; Özbey, S.; Kovalishyn, V.; Dimoglo, A. Synthesis and structure–antituberculosis activity relationship of 1H-indole-2,3-dione derivatives. Bioorg. Med. Chem. 2007, 15, 5888–5904. [Google Scholar] [CrossRef]
  15. Angelova, V.T.; Pencheva, T.; Vassilev, N.; Simeonova, R.; Momekov, G.; Valcheva, V. New indole and indazole derivatives as potential antimycobacterial agents. Med. Chem. Res. 2019, 28, 485–497. [Google Scholar] [CrossRef]
  16. Villanueva, P.J.; Mulia, Y.L.; Sanchez, G.I.; Espinosa, P.J.F.; Arteche, S.O.; Espunes, S.T.D.R.; Cerbon, M.A.; Villar, R.K.; Vicente, R.A.K.; Gines, C.M.; et al. Synthesis and Biological Evaluation of 2H-Indazole Derivatives: Towards Antimicrobial and Anti-Inflammatory Dual Agents. Molecules 2017, 22, 1864. [Google Scholar] [CrossRef] [Green Version]
  17. Lin, Y.C.; Chou, L.C.; Chen, S.C.; Kuo, S.C.; Huang, L.J.; Gean, P.W. Neuroprotective effects of furopyrazole derivative of benzylindazole analogs on C2 ceramide-induced apoptosis in cultured cortical neurons. Bioorg. Med. Chem. Lett. 2009, 19, 3225–3228. [Google Scholar] [CrossRef]
  18. Chen, C.Y.C. De novo design of novel selective COX-2 inhibitors: From virtual screening to pharmacophore analysis. J. Taiwan Inst. Chem. Eng. 2009, 40, 55–69. [Google Scholar] [CrossRef]
  19. Woods, K.W.; Fischer, J.P.; Claiborne, A.; Li, T.; Thomas, S.A.; Zhu, G.-D.; Diebold, R.B.; Liu, X.; Shi, Y.; Klinghofer, V. Synthesis and SAR of indazole-pyridine based protein kinase B/Akt inhibitors. Bioorg. Med. Chem. 2006, 14, 6832–6846. [Google Scholar] [CrossRef]
  20. Yang, L.L.; Chen, Y.; He, J.; Njoya, E.M.; Chen, J.; Liu, S.; Xie, C.; Huang, W.; Wang, F.; Wang, Z.; et al. 4,6-Substituted-1H-Indazoles as potent IDO1/TDO dual inhibitors. Bioorg. Med. Chem. 2019, 27, 1087–1098. [Google Scholar] [CrossRef]
  21. Harada, H.; Morie, T.; Hirokawa, Y.; Terauchi, H.; Fujiwara, I.; Yoshida, N.; Kato, S. Development of potent serotonin-3 (5-HT3) receptor antagonists. II. Structure-activity relationships of N-(1-benzyl-4-methylhexahydro-1H-1, 4-diazepin-6-yl) carboxamides. Chem. Pharm. Bull. 1995, 43, 1912–1930. [Google Scholar] [CrossRef] [Green Version]
  22. Schaus, J.M.; Thompson, D.C.; Bloomquist, W.E.; Susemichel, A.D.; Calligaro, D.O.; Cohen, M.L. Synthesis and Structure− Activity Relationships of Potent and Orally Active 5-HT4 Receptor Antagonists: Indazole and Benzimidazolone Derivatives. J. Med. Chem. 1998, 41, 1943–1955. [Google Scholar] [CrossRef]
  23. Wang, C.; Liu, X.; Xiao, T.; Xu, Z.Q.; Cao, S.; Wang, H.F.; Yan, Q.J.; Gu, S.X.; Zhu, Y.Y. Anticancer activity evaluation of indazolyl-substituted piperidin-4-yl-aminopyrimidines. Med. Chem. Res. 2020, 29, 910–915. [Google Scholar] [CrossRef]
  24. Reddy, S.G.; Mohanty, S.; Kumar, J.; Rao, B.V. Synthesis and Evaluation of Anticancer Activity of Indazole Derivatives. Russ. J. Gen. Chem. 2018, 88, 2394–2399. [Google Scholar] [CrossRef]
  25. Chu, Y.Y.; Cheng, H.J.; Tian, Z.H.; Zhao, J.C.; Li, G.; Chu, Y.Y.; Sun, C.J.; Li, W.B. Rational drug design of indazole-based diarylurea derivatives as anticancer agents. Chem. Biol. Drug Des. 2019, 90, 609–617. [Google Scholar] [CrossRef]
  26. Scott, L.J. Niraparib: First global approval. Drugs 2017, 77, 1029–1034. [Google Scholar] [CrossRef]
  27. Zivi, A.; Cerbone, L.; Recine, F.; Sternberg, C.N. Safety and tolerability of pazopanib in the treatment of renal cell carcinoma. Expert Opin. Drug Saf. 2012, 11, 851–859. [Google Scholar] [CrossRef]
  28. Escudier, B.; Gore, M. Axitinib for the Management of Metastatic Renal Cell Carcinoma. Drugs R D 2011, 11, 113–126. [Google Scholar] [CrossRef]
  29. Egan, B.A.; Burton, P.M. Synthesis of 3-aryl-1 H-indazoles via iridium-catalysed C–H borylation and Suzuki–Miyaura coupling. RSC Adv. 2014, 4, 27726–27729. [Google Scholar] [CrossRef]
  30. Yu, S.; Tang, G.; Li, Y.; Zhou, X.; Lan, Y.; Li, X. Anthranil: An Aminating Reagent Leading to Bifunctionality for Both C (sp3)− H and C (sp2)− H under Rhodium (III) Catalysis. Angew Chem. Int. Ed. 2016, 55, 8696–8700. [Google Scholar] [CrossRef]
  31. Zhu, J.; Sun, S.; Cheng, J. Rh(III)-catalyzed [4 + 1]-annulation of azobenzenes with α- carbonyl sulfoxonium ylides toward 3-acyl-(2H)-indazoles. Tetrahedron Lett. 2018, 59, 2284–2287. [Google Scholar] [CrossRef]
  32. Tang, X.; Gao, H.; Yang, J.; Wu, W.; Jiang, H. Efficient access to 1 H-indazoles via copper-catalyzed cross-coupling/cyclization of 2-bromoaryl oxime acetates and amines. Org. Chem. Front. 2014, 1, 1295–1298. [Google Scholar] [CrossRef]
  33. Hummel, J.R.; Ellman, J.A. Cobalt (III)-catalyzed synthesis of indazoles and furans by C–H bond functionalization/addition/cyclization cascades. J. Am. Chem. Soc. 2014, 137, 490–498. [Google Scholar] [CrossRef] [Green Version]
  34. ang, G.; Sun, J.; Wang, K.; Han, J.; Li, H.S.; Duan, G.; You, G.; Li, F.; Xia, C. Palladium-catalyzed direct C–H nitration and intramolecular C–H functionalization for the synthesis of 3-nitro-1-(phenylsulfonyl)-1H-indazole derivatives. Org. Chem. Front. 2019, 6, 1608–1612. [Google Scholar]
  35. Lebedev, A.Y.; Khartulyari, A.S.; Voskoboynikov, A.Z. Synthesis of 1-aryl-1 H-indazoles via palladium-catalyzed intramolecular amination of aryl halides. J. Org. Chem. 2005, 70, 596–602. [Google Scholar] [CrossRef]
  36. Haag, B.; Peng, Z.; Knochel, P. Preparation of polyfunctional indazoles and heteroarylazo compounds using highly functionalized zinc reagents. Org. Lett. 2009, 11, 4270–4273. [Google Scholar] [CrossRef]
  37. Zhang, T.; Bao, W. Synthesis of 1 H-Indazoles and 1 H-Pyrazoles via FeBr3/O2 Mediated Intramolecular C–H Amination. J. Org. Chem. 2013, 78, 1317–1322. [Google Scholar] [CrossRef]
  38. Liu, Y.L.; Pan, Y.L.; Li, G.J.; Xu, H.F.; Chen, J.Z. The nickel-catalyzed C3-acylation of 2H-indazoles with aldehydes. Org. Biomol. Chem. 2019, 17, 8749–8755. [Google Scholar] [CrossRef]
  39. Boddapati, S.N.M.; Saketi, J.M.R.; Mutchu, B.R.; Bollikolla, H.B.; Adil, S.F.; Khan, M. Copper Promoted Desulfurization and C-N Cross Coupling Reactions: Simple approach to the synthesis of substituted 2-Aminobenzoxazoles and 2,5-Disubstituted Tetrazole amines. Arab. J. Chem. 2020, 13, 4477–4494. [Google Scholar] [CrossRef]
  40. Boddapati, S.N.M.; Tamminana, R.; Gollapudi, R.K.; Nurbasha, S.; Assal, M.E.; Alduhaish, O.; Siddiqui, M.R.H.; Bollikolla, H.B.; Adil, S.F. Copper-Promoted One-Pot Approach: Synthesis of Benzimidazoles. Molecules 2020, 25, 1788. [Google Scholar] [CrossRef] [Green Version]
  41. Boddapati, S.N.M.; Kurmarayuni, C.M.; Mutchu, B.R.; Tamminana, R.; Bollikolla, H.B. Copper-catalyzed synthesis of 2-aminophenyl benzothiazoles: A novel approach. Org. Biomol. Chem. 2018, 16, 8267–8272. [Google Scholar] [CrossRef]
  42. Boddapati, S.N.M.; Kola, A.E.; Kesana, S.B.; Bollikolla, H.B. Temperature dependent regioselective synthesis of aryl tetrazole amines using copper source. J. Organomet. Chem. 2018, 866, 177–183. [Google Scholar] [CrossRef]
  43. Boddapati, S.N.M.; Polam, N.; Mutchu, B.R.; Bollikolla, H.B. The synthesis of arylcyanamides: A copper catalyzed consecutive desulfurization and C-N cross coupling strategy. New J. Chem. 2018, 42, 918–922. [Google Scholar] [CrossRef]
  44. Reddy, O.S.; Suryanarayana, C.V.; Narayana, K.; Anuradha, V.; Babu, B.H. Synthesis and cytotoxic evaluation for some new 2,5-disubstituted pyrimidine derivatives for anticancer activity. Med. Chem. Res. 2015, 24, 1777–1788. [Google Scholar]
  45. Siva Nagi Reddy, M.; Sailaja Kumari, B.; Hari Babu, B. Synthesis and Cytotoxicity Evaluation of Some Novel 3,5-disubstituted-1, 2,4-Oxadiazoles. Lett. Drug. Des. Discov. 2012, 9, 942–946. [Google Scholar] [CrossRef]
  46. Mutchu, B.R.; Kotra, V.; Onteddu, S.R.; Boddapati, S.N.M.; Bollikolla, H.B. Synthesis, Cytotoxicity and Antimicrobial Evaluation of Some New 2-Aryl, 5-Substituted 1,3,4-Oxadiazoles and 1,3,4-Thiadiazoles. Chem. Afr. 2019, 2, 15–20. [Google Scholar]
  47. Scudiero, D.A.; Shoemaker, R.H.; Paull, K.D.; Monks, A.; Tierney, S.; Nofziger, T.H.; Currens, M.J.; Seniff, D.; Boyd, M.R. Evaluation of a soluble tetrazolium/formazan assay for cell growth and drug sensitivity in culture using human and other tumor cell lines. Cancer Res. 1988, 48, 4827–4833. [Google Scholar]
  48. Barile, E.; De, S.K.; Carlson, C.B.; Chen, V.; Knutzen, C.; Mehan, M.R.; Yang, L.; Dahl, R.; Chiang, G.; Pellecchia, M. Design, Synthesis, and Structure−Activity Relationships of 3-Ethynyl-1H-indazoles as Inhibitors of the Phosphatidylinositol 3-Kinase Signaling Pathway. J. Med. Chem. 2010, 53, 8368–8375. [Google Scholar]
  49. Atobe, M.; Naganuma, K.; Kawanishi, M.; Morimoto, A.; Kasahara, K.I.; Ohashi, S.; Suzuki, H.; Hayashi, T.; Miyoshi, S. Discovery of 2-(1H-indazol-1-yl)-thiazole derivatives as selective EP1 receptor antagonists for treatment of overactive bladder by core structure replacement. Bioorg. Med. Chem. Lett. 2014, 24, 1327–1333. [Google Scholar] [CrossRef]
  50. Kusakabe, K.; Ide, N.; Daigo, Y.; Tachibana, Y.; Itoh, T.; Yamamoto, T.; Hashizume, H.; Hato, Y.; Higashino, K.; Okano, Y.; et al. Indazole-Based Potent and Cell-Active Mps1 Kinase Inhibitors: Rational Design from Pan-Kinase Inhibitor Anthrapyrazolone (SP600125). J. Med. Chem. 2013, 56, 4343–4356. [Google Scholar] [CrossRef]
  51. Pelc, M.; Huang, W.; Trujillo, J.; Baldus, J.; Turner, S.; Kleine, P.; Yang, S.; Thorarensen, A. An efficient and regioselective difluoromethylation of 3-iodoindazole with chlorodifluoromethane. Synlett 2010, 2010, 219–222. [Google Scholar] [CrossRef]
  52. Xiao, J.; Jin, C.; Liu, Z.; Guo, S.; Zhang, X.; Zhou, X.; Wu, X. The design, synthesis, and biological evaluation of novel YC-1 derivatives as potent anti-hepatic fibrosis agents. Org. Biomol. Chem. 2015, 13, 7257–7264. [Google Scholar]
  53. Hunt, K.W.; Moreno, D.A.; Suiter, N.; Clark, C.T.; Kim, G. Selective Synthesis of 1-Functionalized-alkyl-1H-indazoles. Org. Lett. 2009, 11, 5054–5057. [Google Scholar] [CrossRef]
Figure 1. Some biologically active molecules of indazoles.
Figure 1. Some biologically active molecules of indazoles.
Applsci 10 03792 g001
Figure 2. Indazoles containing anticancer drugs.
Figure 2. Indazoles containing anticancer drugs.
Applsci 10 03792 g002
Scheme 1. Synthetic route for N-methyl-3-aryl indazoles.
Scheme 1. Synthetic route for N-methyl-3-aryl indazoles.
Applsci 10 03792 sch001
Figure 3. Illustration of the IC50 values for the compounds 3(a–ja; b; c; d; e; f; g; h; i; j) and 5(a–ja; b; c; d; e; f; g; h; i; j; a; b; c; d; e; f; g; h; i; j).
Figure 3. Illustration of the IC50 values for the compounds 3(a–ja; b; c; d; e; f; g; h; i; j) and 5(a–ja; b; c; d; e; f; g; h; i; j; a; b; c; d; e; f; g; h; i; j).
Applsci 10 03792 g003
Table 1. Details of the series of compounds 3a–3j.
Table 1. Details of the series of compounds 3a–3j.
CompoundR-B(OH)2
R =
Product
3a–3j
Time (h)Yield (%)M.P. (°C)
3a Applsci 10 03792 i001 Applsci 10 03792 i0021265114–116
3b Applsci 10 03792 i003 Applsci 10 03792 i0041067136–138
3c Applsci 10 03792 i005 Applsci 10 03792 i006955112–113
3d Applsci 10 03792 i007 Applsci 10 03792 i008863101–103
3e Applsci 10 03792 i009 Applsci 10 03792 i010862184–186
3f Applsci 10 03792 i011 Applsci 10 03792 i01297085–77
3g Applsci 10 03792 i013 Applsci 10 03792 i0141070123–125
3h Applsci 10 03792 i015 Applsci 10 03792 i016968115–116
3i Applsci 10 03792 i017 Applsci 10 03792 i018860121–123
3j Applsci 10 03792 i019 Applsci 10 03792 i0201160116–117
Table 2. Details of the series of compounds 5a–5j.
Table 2. Details of the series of compounds 5a–5j.
EntryR-B(OH)2
R =
Product
5a–5j
Time (h)Yield (%)M.P. (°C)
5a Applsci 10 03792 i001 Applsci 10 03792 i021127076–78
5b Applsci 10 03792 i003 Applsci 10 03792 i0221172137–139
5c Applsci 10 03792 i005 Applsci 10 03792 i023145898–100
5d Applsci 10 03792 i007 Applsci 10 03792 i0241072101–103
5e Applsci 10 03792 i009 Applsci 10 03792 i025117074–76
5f Applsci 10 03792 i011 Applsci 10 03792 i0261075112–114
5g Applsci 10 03792 i013 Applsci 10 03792 i027107399–101
5h Applsci 10 03792 i015 Applsci 10 03792 i0281172105–107
5i Applsci 10 03792 i017 Applsci 10 03792 i0291069241–243
5j Applsci 10 03792 i019 Applsci 10 03792 i0301267110–112
Table 3. Anticancer activity of compounds 3a–3j.
Table 3. Anticancer activity of compounds 3a–3j.
CompoundIC50 (μg/mL)
HCT-116MDA-MB-231
3a----
3b----
3c92.9 ± 6.5102.3 ± 13.2
3d127.6 ± 3.8117.5 ± 15.0
3e----
3f109.6 ± 12.3107.4 ± 10.0
3g106.6 ± 14112.2 ± 17.9
3h93.6 ± 7.295 ± 6.8
3i102 ± 11.1120.1 ± 7.2
3j85.3 ± 16.787 ± 5.2
Standard *1.2 ± 0.30.3 ± 0.1
“--” indicates IC50 value >200 μg/mL; IC50 values are reported in micromolar concentrations of the compound to affect 50% inhibition of the tumor cell growth; * doxorubicin is employed as standard.
Table 4. Anticancer activity of compounds 5a–5j.
Table 4. Anticancer activity of compounds 5a–5j.
CompoundIC50 (μg/mL)
HCT-116MDA-MB-231
5a148.8 ± 4.7172.4 ± 14.2
5b110.6 ± 5.9125.7 ± 13.1
5c63.7 ± 14.058.2 ± 14.7
5d62.3±15.081.0±3.4
5e102 ± 14.0150.1 ± 7.2
5f69.0 ± 18.080.7 ± 10.5
5g72.7 ± 20.088.5 ± 10.1
5h90.1 ± 1.598.6 ± 18
5i62.3 ± 6.079.0 ± 7.2
5j79.4 ± 7.487.8 ± 6.9
Standard *1.2 ± 0.30.3 ± 0.2
“--” indicates IC50 value >200 μg/mL; IC50 values are reported in micromolar concentrations of the compound to affect 50% inhibition of the tumor cell growth; * doxorubicin is employed as standard.

Share and Cite

MDPI and ACS Style

Saketi, J.M.R.; Boddapati, S.N.M.; M., R.; Adil, S.F.; Shaik, M.R.; Alduhaish, O.; Siddiqui, M.R.H.; Bollikolla, H.B. Pd(PPh3)4 Catalyzed Synthesis of Indazole Derivatives as Potent Anticancer Drug. Appl. Sci. 2020, 10, 3792. https://doi.org/10.3390/app10113792

AMA Style

Saketi JMR, Boddapati SNM, M. R, Adil SF, Shaik MR, Alduhaish O, Siddiqui MRH, Bollikolla HB. Pd(PPh3)4 Catalyzed Synthesis of Indazole Derivatives as Potent Anticancer Drug. Applied Sciences. 2020; 10(11):3792. https://doi.org/10.3390/app10113792

Chicago/Turabian Style

Saketi, Jagan Mohana Rao, S. N. Murthy Boddapati, Raghuram M., Syed Farooq Adil, Mohammed Rafi Shaik, Osamah Alduhaish, Mohammed Rafiq H. Siddiqui, and Hari Babu Bollikolla. 2020. "Pd(PPh3)4 Catalyzed Synthesis of Indazole Derivatives as Potent Anticancer Drug" Applied Sciences 10, no. 11: 3792. https://doi.org/10.3390/app10113792

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop