You are on page 1of 12

Med Chem Res (2017) 26:929–939 MEDICINAL

DOI 10.1007/s00044-017-1798-9
CHEMISTRY
RESEARCH
ORIGINAL RESEARCH

Design, synthesis and biological evaluation of new 4-(4-substituted-


anilino)quinoline derivatives as anticancer agents
Khaled R. A. Abdellatif1,2 Eman K. A. Abdelall1 Mohamed A. Abdelgawad1,3
● ● ●

Dina M. E. Amin1 Hany A. Omar4,5


Received: 7 September 2016 / Accepted: 3 February 2017 / Published online: 23 February 2017
© Springer Science+Business Media New York 2017

Abstract A new series of 4-(4-substituted-anilino)quino- Introduction


line derivatives 6a–f was synthesized from amine deriva-
tives via Gould–Jacobs reaction. All synthesized The quinoline scaffold, a highly versatile drug like template,
compounds were evaluated for their cytotoxic activity is often used for the design of many pharmaceutical active
against two human cancer cell lines; breast carcinoma compounds with different pharmacological activities such
(MCF-7) and non-small cell lung cancer (A549). The tested as antimalarial (Ekoue et al. 2009), antimicrobial (Patel and
compounds showed a broad range of activities (IC50 = Park 2014), anti-inflammatory (El-Feky et al. 2015), hyp-
3.42–23.32 and 5.97–22.01 µM) in comparison with dox- notic (Ferlin et al. 2005), ant tuberculosis (Medapi et al.
orubicin (IC50 = 2.07 and 0.02 µM) and erlotinib (IC50 = 2015), anticancer (Sun et al. 2013), antiviral (Majerz et al.
1.14 and 19.26 µM) for MCF-7 and A549 respectively. The 2011), antischizophrenic (Hamaguchi et al. 2015), anti-
4-(4-chloroanilino)quinoline derivative (6c, IC50 = 3.42 and protozoal (Ma et al. 2009), anticholesterol (Rano et al.
5.97 µM) was the most potent among all compounds against 2009), Anthelmintic (Rossiter et al. 2005), and analgesic
both MCF-7 and A549 cell lines respectively. In addition, (Roma et al. 2008). Quinoline is a privileged anticancer
molecular docking studies were performed and the results scaffold possessing various antiproliferative mechanisms as
were in agreement with the in vitro cytotoxic data. cell cycle arrest, angiogenesis inhibition, apoptosis (Afzal
et al. 2015), DNA repair and tyrosine kinases (TK) inhibitor
Keywords Quinoline Antitumor MCF-7 EGFR
● ● ●
(Solomon and Lee 2011). Epidermal growth factor receptor
(EGFR), one of protein kinases, plays a critical role in
signals transduction (Abdellatif et al. 2014a). Dysregulation
of EGFR causes considerable alteration in different cellular
processes like transcription, proliferation, angiogensis and
inhibition of apoptosis so drugs targeted EGFR are con-
* Khaled R. A. Abdellatif sidered as the main anticancer chemotherapeutics (Hicks
khaled.ahmed@pharm.bsu.edu.eg et al. 2010) especially for breast, ovarian, colon, lung, and
1 prostate cancer at which over expression of EGFR was
Department of Pharmaceutical Organic Chemistry, Faculty of
Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt established (Amin et al. 2011). Erlotinib (I) is one of 4-
2 anilinoqunazoline derivatives approved by FDA as kinase
Pharmaceutical Sciences Department, Ibn Sina National College
for Medical Studies, Jeddah 21418, Saudi Arabia inhibitor (Arora and Scholar 2005), 6,7-disubstituted-4-
3 anilinoquioline-3-carbonitrile derivatives EKB-567 (II) and
Department of Pharmaceutical Chemistry, Faculty of Pharmacy,
Al-Jouf University, Sakaka, Al-Jouf, Saudi Arabia HKI-272 (III) are EGFR inhibitors which were selected for
4 further studies in clinical trials as anticancer (Afzal et al.
Department of Pharmacology and Toxicology, Faculty of
Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt 2015) were taken as references in our design for new
5 cytotoxic quinoline derivatives (Fig. 1).
Sharjah Institute for Medical Research, Department of
Pharmacology, College of Pharmacy, University of Sharjah, Based on the previous information and in continuation of
Sharjah 27272, United Arab Emirates our previous work for synthesis of new anticancer agents

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


930 Med Chem Res (2017) 26:929–939

Fig. 1 Rational design of 4-(4-


substituted-anilino)quinoline
derivatives 6a–f based on
chemical structures of some
anticancer drugs containing
anilinoquinazoline and
anilinoquinoline moieties
(erlotinib I, EKB-569 II and
HKI-272 III) (Arora and
Scholar 2005; Afzal et al. 2015)

(Abdellatif et al. 2013, 2014b; Gouda et al. 2014), we now interaction and (iv) replacement the large fragment of the
describe synthesis, in vitro anticancer evaluation and erlotinib, aminoacrylate and alkoxy moieties in positions
molecular docking for a new series of 4-anilinoquinoline 6,7 of EKB-569 and HKI-272 quinoline rings by electron
derivatives 6a–f which are structurally related to erlotinib withdrawing groups as COOH or Br.
(I), EKB-567 (II) and HKI-272 (III) with some modifica-
tions; (i) isosteric replacement of quniazoline ring of erlo-
tinib by quinoline ring which bind with adenine portion of Material and methods
ATP binding site of EGFR protein tyrosine kinase, (ii)
attach different substituents at para position of the aniline Experimental
moiety (lipophilic group as methyl or methoxy, 6a, 6b, 6d
and 6e or halogen like Cl, 6c and 6f) to check their effect on Melting points were determined on a Griffin apparatus and
biological activity, (iii) the N of quniazoline ring of erloti- are uncorrected. Infrared (IR) spectra were recorded on
nib (I) or the cyano group at position 3 of quinoline ring for Shimadzu IR 435 spectrophotometer using KBr discs. 1H
both EKB-569 (II) and HKI-272 (III) were replaced by NMR spectra were measured on Varian Gemini 300 MHz or
another electron withdrawing ester group to retain the on Bruker 400 MHz spectrometer in D2O, CDCl3 or
charge distribution that would result from such EGFR DMSO-d6 with TMS as the internal standard, where J

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Med Chem Res (2017) 26:929–939 931

(coupling constant) values were estimated in hertz (Hz) and Anal. Calcd. for C15H16BrNO6 (386): C, 46.65; H, 4.18; N,
chemical shifts were recorded in ppm on δ scale. 13C NMR 3.63. Found: C, 46.88; H, 4.20; N, 3.63.
spectra were carried out on Bruker 400 MHz spectrometer.
Mass spectra were run at 70 ev on Shimadzu QP-2010 plus General procedure for synthesis of quinoline compounds
spectrometer. Microanalyses were performed for C, H, N (4a, b)
(Micro analytical Center, Cairo University and at the
regional center for Mycology and Biotechnology, Al-Azhar A solution of 3a or 3b (0.772 g, 2.0 mmol) in diphenyl ether
University) and were within +0.4% of theoretical values. (30 mL) was heated at 250–260 °C for 1–2 h .The reaction
Progress of the reaction was monitored by TLC using alu- mixture was cooled and then diethyl ether (100 mL) was
minum sheets precoated with UV fluorescent silica gel added. The resulting precipitate was filtered, washed with
Merck 60 F254 and were visualized by UV lamp. petroleum ether followed by methanol, dried and crystal-
lized from ethanol. Physical and spectral data of 4a, b are
General procedure for synthesis of enamine compounds listed below.
(3a, b)
8-Bromo-3-(ethoxycarbonyl)-4-oxo-1,4-dihydroquinoline-
A mixture of the respective amine1a or 1b (2.16 g, 10 6-carboxylic acid (4a) Shiny yellow crystals; yield 63%;
mmol) with diethylethoxymethylene malonate (2, 2.16 g, m.p. 293–295 °C; IR (KBr/cm−1) 3446 (OH), 3129 (NH),
10 mmol) in ethanol (25 mL) was heated under reflux for 3090 (C–H aromatic), 2980 (CH aliphatic), 1688 (C=O),
5–7 h. The mixture was evaporated under reduced pressure 1633 (C=O); 1H NMR (DMSO-d6) δ 1.29 (t, J = 6.9 Hz,
to give a solid product which crystallized from benzene. 3H, CH2CH3), 4.24 (q, J = 6.9 Hz, 2H, CH2CH3), 8.39 (s,
Physical and spectral data of 3a, b are listed below. 1H, quinoline H-2), 8.46 (s, 1H, quinoline H-5), 8.70 (s,
1H, quinoline H-7), 11.56 (s, 1H, NH, D2O exchangeable),
3-bromo-4-((3-ethoxy-2-(ethoxycarbonyl)-3-oxoprop-1-en- 13.30 (s, 1H, OH, D2O exchangeable); 13C NMR (DMSO-
1-yl)amino)benzoic acid (3a) White crystals; yield 83%; d6) δ 14.5, 60.83, 111.7, 112.6, 127.6, 128.2, 136.2, 137.9,
m.p. 222–224 °C; IR (KBr/cm−1) 3486 (OH), 3385 (NH), 143.2, 146.5, 164.4, 166.1; MS, (relative abundance %):
þ þ
3090 (C–H aromatic), 2983 (C–H aliphatic), 1692 (C=O), 340 (M  , 37.50 %), 342 (M  +2, 35.57%), 341 (100%);
1627 (C=O); 1H NMR (CDCl3) δ 1.21–1.27 (m, 6H, 2 Anal. Calcd. for C13H10BrNO5 (340): C, 45.91; H, 2.96; N,
(CH2CH3)), 4.17 (q, J = 7.2 Hz, 2H, CH2CH3), 4.26 (q, J 4.12. Found: C, 45.70; H, 3.00; N, 4.06.
= 7.2 Hz, 2H, CH2CH3), 7.72 (d, J = 8.7 Hz, 1H, phenyl H-
6), 7.95 (d, J = 8.7 Hz, 1H, phenyl H-5), 8.14 (s, 1H, 6-Bromo-3-(ethoxycarbonyl)-4-oxo-1,4-dihydroquinoline-
phenyl H-3), 8.54 (d, J = 12.6 Hz, 1H, HC olefenic), 11.21 8-carboxylic acid (4b) Shiny light green crystals; yield
(d, J = 12.6 Hz, 1H, NH, D2O exchangeable), 13.30 ppm (s, 60%; m.p. 288–290 °C; IR (KBr/cm−1) 3475 (OH), 3431
1H, OH, D2O exchangeable); 13C NMR (CDCl3) δ 14.2, (NH), 3076 (C–H aromatic), 2984 (CH aliphatic), 1690
14.3, 60.6, 60.9, 97.8, 112.9, 114.5, 119.2, 128.8, 130.7, (C=O), 1648 (C=O); 1H NMR (DMSO-d6) δ 1.30 (t, J =
135.5, 146.1, 149.1, 168.5, 170.2; MS, (relative abundance 7.2 Hz, 3H, CH2CH3), 4.23 (q, J = 7.2 Hz, 2H, CH2CH3),
þ þ
%) 386 (M  , 9.63 %), 388 (M  +2, 8.87%), 341 (100%); 8.38 (s, 1H, quinoline H-2), 8.69 (s,1H, quinoline H-5),
Anal. Calcd. for C15H16BrNO6 (386): C, 46.65; H, 4.18; N, 8.70 (s, 1H, quinoline H-7), 11.65 (s, 1H, NH, D2O
3.93. Found: C, 46.75; H, 3.88; N, 4.07. exchangeable), 13.32 (s,1H, OH, D2O exchangeable); 13C
NMR (DMSO-d6)δ 14.7, 60.1, 111.0, 113.0, 127.67, 127.9,
5-Bromo-2-((3-ethoxy-2-(ethoxycarbonyl)-3-oxoprop-1-en- 128.9, 135.2, 143.2, 147.2, 164.4, 165.8, 173.4; MS,
þ þ
1-yl)amino)benzoic acid (3b) Shiny buff crystals; yield (relative abundance %): 340 (M  , 33.66 %), 342 (M  +2,
80%; m.p. 198–200 °C; IR (KBr/cm−1) 3465 (OH), 3130 31.71), 341 (100%); Anal. Calcd. for C13H10BrNO5
(NH), 3088 (C–H aromatic), 2981 (C–H aliphatic), 1688 (340): C, 45.91; H, 2.96; N, 4.12. Found: C, 45.81; H, 3.02;
(C=O), 1633 (C=O); 1H NMR (CDCl3) δ 1.35–1.43 (m, N, 3.98.
6H, 2(CH2CH3)), 4.30 (q, J = 6.8 Hz, 2H, CH2CH3), 4.38
(q, J = 6.8 Hz, 2H, CH2CH3), 7.36 (d, J = 8.4 Hz, 1H, General procedure for synthesis of 4-chloroquinoline
phenyl H-6), 8.10 (d, J = 8.4 Hz, 1H, phenyl H-5), 8.36 (s, compounds (5a, b)
1H, phenyl H-3), 8.55 (d, J = 12.8 Hz, 1H, HC olefenic),
11.46 (d, J = 12.8 Hz, 1H, NH, D2O exchangeable), 13.25 A quinoline derivative 4a or 4b (1.02 g, 3.0 mmol) was
(s, 1H, OH, D2O exchangeable ); 13C NMR (CDCl3) δ 14.3, added to POCl3 (2.29 g, 15.0 mmol), and the mixture was
14.3, 60.6, 60.7, 97.6, 106.5, 111.2, 115.6, 116.6, 117.2, heated under reflux until the starting material had dis-
133.8, 135.1, 148.6, 168.5, 170.2; MS, (relative abundance appeared at TLC (8 h). The suspension was then cooled first
þ þ
%): 386 (M  , 8.47 %), 388 (M  +2, 7.57%), 115 (100%); to room temperature and then to 0 °C in a water/ice bath.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


932 Med Chem Res (2017) 26:929–939

Then, with stirring it was carefully made alkaline with quinoline H-7), 8.47 (d, J = 12.9 Hz, quinoline H-2), 11.37
Na2CO3 aqueous solution, then this filtrate was acidified by (s, J = 12.9 Hz, 1H, NH ,D2O exchangeable );13C NMR
acetic acid (96%) and the resulting precipitate was col- (CDCl3) δ14.2, 20.9, 61.4, 106.0, 119.2, 123.7, 125.0,
lected, washed many times with water, dried and crystal- 125.7, 129.1, 130.4, 134.1, 135.9, 139.8, 150.1, 153.9,
þ
lized from methanol. Physical and spectral data of 5a, b are 154.1, 164.5, 168.1; MS, (relative abundance %): 429 (M  ,
þ
listed below. 6.70 %), 431(M  +2, 6.16%), 322(100%); Anal. Calcd. for
C20H17BrN2O4 (429): C, 55.96; H, 3.99; N, 6.53. Found: C,
8-Bromo-4-chloro-3-(ethoxycarbonyl)quinoline-6-car- 55.60; H, 3.70; N, 6.70.
boxylic acid (5a) Canary yellow crystals; yield 73%; m.p.
244–246 °C; IR (KBr/cm−1) 3436 (OH), 3089 (C–H aro- 8-Bromo-3-(ethoxycarbonyl)-4-((4-methoxyphenyl)amino)
matic), 2983 (CH aliphatic), 1691 (C=O), 1653 (C=O); 1H quinoline-6-carboxylic acid (6b) Yellow crystals; yield
NMR (DMSO-d6 ) δ 1.28 (t, J = 7.2 Hz, 3H, CH2CH3), 69%; m.p. 182–184 °C; IR (KBr/cm−1) 3421–3272 (OH,
4.23 (q, J = 7.2 Hz, 2H, CH2CH3), 8.37 (s, 1H, quinoline NH), 3090 (C–H aromatic), 2928 (C–H aliphatic), 1716
H-7), 8.44 (s, 1H, quinoline H-5), 8.69 (s, 1H, quinoline H- (C=O); 1H NMR (CDCl3) δ 1.26 (t, J = 7.2 Hz, 3H,
2), 11.85 (s, 1H, OH, D2O exchangeable); 13C NMR CH2CH3), 3.80 (s, 3H, OCH3), 4.46 (q, J = 7.2 Hz, 2H,
(DMSO-d6) δ 14.7, 60.1, 111.3, 127.6, 128.2, 128.6, 135.2, CH2CH3), 6.91 (d, J = 8.8 Hz, 2H, phenyl H-2,H-6), 7.10
147.5, 164.4, 166.1, 173.4; MS, (relative abundance %): (d, J = 8.8 Hz, 2H , phenyl H-3,H-5), 8.43 (s, 1H, quinoline
þ þ
358.5 (M  , 52.35%), 360.5 (M  +2, 14.79%), 57(100%); H-5), 8.53 (s, 1H, quinoline H-7), 9.42 (s, 1H, quinoline H-
Anal. Calcd. for C13H9BrClNO4 (358.5): C, 43.54; H, 2.53; 2), 10.83 (s, 1H, NH, D2O exchangeable);13C NMR
N, 3.91. Found: C, 43.28; H, 2.79; N, 4.13. (CDCl3) δ 14.2, 55.4, 61.4, 105.3, 115.1, 118.6, 124.2,
125.6, 129.0, 134.3, 149.5, 153.9, 154.7, 158.3, 164.1,
þ
6-Bromo-4-chloro-3-(ethoxycarbonyl)quinoline-8-car- 168.2; MS, (relative abundance %): 445 (M  12.07 %), 447
þ
boxylic acid (5b) Yellow crystals; yield 70%; m.p. (M  +2, 10.82), 278 (100%); Anal. Calcd. for
215–217 °C; IR (KBr/cm−1) 3426 (OH), 3076 (C–H aro- C20H17BrN2O5 (445): C, 53.95; H, 3.85; N, 6.29. Found: C,
matic) , 2980 (CH aliphatic), 1756 (C=O), 1713 (C=O); 1H 54.06; H, 3.91; N, 6.40.
NMR (DMSO-d6) δ 1.29 (t, J = 6.8 Hz, 3H, CH2CH3), 4.24
(q, J = 6.8 Hz, 2H, CH2CH3), 8.38 (s, 1H, quinoline H-5), 8-Bromo-4-((4-chlorophenyl)amino)-3-(ethoxycarbonyl)
8.45 (s,1H, quinoline H-7), 8.69 (s, 1H, quinoline H-2), quinoline-6-carboxylic acid (6c) Yellow crystals; yield
11.91 (s, 1H, OH, D2O exchangeable); MS, (relative 71%; m.p. 200–202 °C ; IR (KBr/cm−1) 3430–3296 (OH,
þ þ
abundance %), 358.5 (M  , 25.35 %), 360.5 (M  +2, NH), 3124 (C–H aromatic), 2982 (C–H aliphatic), 1715
15.21%), 75 (100%); Anal. Calcd. for C13H9BrClNO4 (C=O); 1H NMR (DMSO-d6) δ 1.13 (t, J = 7.2 Hz, 3H,
(358.5): C, 43.54; H, 2.53; N, 3.91. Found: C, 43.64; H, CH2CH3) , 3.95 (q, J = 7.2 Hz, 2H, CH2CH3), 7.12 (d, J =
2.90; N, 3.73. 8.8 Hz, 2H, phenyl H-2,H-6), 7.36 (d, J = 8.8 Hz, 2H,
phenyl H-3,H-5), 8.49 (s, 1H, quinoline H-5), 8.90 (s, 1H,
General procedure for synthesis of 4-substituted quinoline H-7), 9.01 (s, 1H, quinoline H-2), 10.03 (s, 1H,
phenylaminoquinoline compounds (6a–f) NH,D2O exchangeable); 13C NMR (CDCl3) δ16.7, 64.2,
109.5, 121.6, 127.1, 127.9, 128.0, 131.2, 132.8, 133.8,
A mixture of chloroquinoline derivative 5a or 5b (3.585 g, 136.8, 142.97, 152.5, 155.9, 156.3, 166.9, 170.4; MS,
þ þ
10 mmol), the appropriate aromatic amine (10 mmol) and (relative abundance %):449 (M  , 3.61%), 451(M  +2,
catalytic amount of sodium iodide (0.2 g) in ethanol (20 6.20%), 322 (100%); Anal. Calcd. for C19H14BrClN2O4
mL) was heated under reflux for 2–4 h. Filter, the filtrate (449): C, 50.75; H, 3.14; N, 6.23. Found: C, 50.55; H, 2.93;
was neutralized to litmus paper with acetic acid (96%). The N, 6.01.
formed precipitate was collected by filtration, washed with
water and crystallized from the ethanol. 6-Bromo-3-(ethoxycarbonyl)-4-(p-tolylamino)quinoline-8-
carboxylic acid (6d) Yellow crystals; yield 62%; m.p.
8-Bromo-3-(ethoxycarbonyl)-4-(p-tolylamino)quinoline-6- 206–208 °C; IR (KBr/cm−1) 3500–3290 (OH, NH), 3100
carboxylic acid (6a) Yellow crystals; yield 65%; m.p. (C–H aromatic), 2975 (C–H aliphatic), 1700 (C=O); 1H
196–198 °C; IR (KBr/cm−1) 3418–3272 (OH, NH), 3127 NMR (CDCl3) δ 1.39 (t, J = 7.6 Hz, 3H, CH2CH3), 2.78 (s,
(C–H aromatic), 2981 (C–H aliphatic), 1705 (C=O); 1H 3H, CH3), 4.30 (q, J = 7.6 Hz, 2H, CH2CH3), 7.21 (d, J =
NMR (CDCl3) δ 1.36 (t, J = 7.2 Hz, 3H, CH2CH3), 2.35 (s, 8.4 Hz, 2H, phenyl H-2,H-6), 7.53 (d, J = 8.4 Hz, 2H,
3H, CH3), 4.48 (q, J = 7.2 Hz, 2H, CH2CH3), 7.17 (d, J = phenyl H-3,H-5), 8.09 (s, 1H, quinoline H-5), 8.35 (s, 1H,
8.4 Hz, 2H, phenyl H-2,H-6), 7.27 (d, J = 8.4 Hz, 2H, quinoline H-7), 8.66 (s, 1H, quinoline H-2), 11.43 (s, 1H,
phenyl H-3,H-5), 8.41 (s, 1H, quinoline H-5), 8.52 (s, 1H, NH ,D2O exchangeable ); 13C NMR (DMSO-d6) δ 18.7,

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Med Chem Res (2017) 26:929–939 933

20.9, 56.5, 120.5, 122.3, 125.0, 127.0, 128.6, 130.5, 133.7, log-phase growth were harvested by trypsinization for use
134.4, 140.2, 149.2, 152.9, 154.4, 165.9, 169.4; MS, in various assays.
þ þ
(relative abundance %): 429 (M  , 14.62 %), 431(M  +2,
12.53%), 64 (100%); Anal. Calcd. for C20H17BrN2O4 Measurement of potential cytotoxic activity
(429): C, 55.96; H, 3.99; N, 6.53. Found: C, 56.07; H, 4.06;
N, 6.70. Cell viability was assessed using MTT assay (Sigma-
Aldrich, St. Louis, MO, USA) in 6 replicates as described
6-Bromo-3-(ethoxycarbonyl)-4-((4-methoxyphenyl)amino) previously (Omar et al. 2013). In brief, cells were seeded
quinoline-8-carboxylic acid (6e) Buff crystals; yield 69%; into 96-well tissue culture plates in DMEM containing 10%
m.p. 203–205 °C; IR (KBr/cm−1) 3440–3208 (OH, NH), FBS to a final volume of 0.2 mL. The cells were subjected
3090 (C–-H aromatic), 2931 (C–H aliphatic), 1710 (C=O); to different treatments 24 h post seeding. Following the
1
H NMR (DMSO-d6) δ 2.5 (t, J = 7.2 Hz, 3H, CH2CH3), incubation for 48 h with doxorubicin (positive control), test
3.10 (s, 3H, OCH3), 3.76 (q, J = 7.2 Hz, 2H,CH2CH3), 6.91 drugs or vehicle (DMSO), the media were removed,
(d, J = 8.8 Hz, 2H, phenyl H-2,H-6 ), 7.10 (d, J = 8.8 Hz, replaced by 200 µL DMEM containing 0.5 mg/mL of MTT
2H , phenyl H-3,H-5), 8.33 (s, 1H, quinoline H-5), 8.39 (s, and cells were incubated for 2 h. Next, the supernatants
1H, quinoline H-7), 9.18 (s, 1H, quinoline H-2), 11.36 (s, were removed and the precipitated formazan was dissolved
1H, NH, D2O exchangeable); 13C NMR (DMSO-d6) δ 22.0, by adding 200 μL of DMSO. Absorbance at 570 nm was
48.8, 79.4, 110.3, 120.9, 121.0, 123.3, 127.7, 128.0, 128.5, determined using a microplate reader (Model 450 Mior-
129.7, 134.32, 141.9, 151.9, 154.1, 165.8, 168.8; MS, oplate Reader; Bio-Rad). Results were calculated by sub-
þ þ tracting blank readings. The relation between surviving
(relative abundance %): 445 (M  , 6.37 %), 447(M  +2,
5.77%), 123 (100%); Anal. Calcd. for C20H17BrN2O5 fraction and drug concentration is plotted and IC50 [the
(445): C, 53.95; H, 3.85; N, 6.29. Found: C, 53.70; H, 4.13; concentration required for 50% inhibition of cell viability]
N, 6.14. was calculated for each compound.
The results of in vitro cytotoxic activity experiments are
presented in (Table 1).
6-Bromo-4-((4-chlorophenyl)amino)-3-(ethoxycarbonyl)
quinoline-8-carboxylic acid (6f) Yellow crystals; yield Docking study
70%; m.p. 211–213 °C; IR (KBr/cm−1) 3434 (NH), 3075
(C–H aromatic), 2977 (C–H aliphatic), 1678 (C=O); 1 H All molecular calculations and docking studies were per-
NMR (DMSO-d6) δ 2.27 (t, J = 7.2 Hz, 3H, CH2CH3), 2.90 formed using “molecular operating environment” (MOE)
(q, J = 7.2 Hz, 2H, CH2CH3), 6.87 (d, J = 8.8 Hz, 2H, version 2008.10 release of Chemical Computing Group’s.
phenyl H-2,H-6), 7.07 (d, J = 8.8 Hz, 2H, phenyl H-3,H-5), The program operated under “Windows XP” operating
8.17 (s, 1H, quinoline H-5), 8.23 (s, 1H, quinoline H-7), system installed on an Intel Pentium IV PC with a 2.8 MHz
8.95 (s, 1H, quinoline H-2), 10.66 (s, 1H, NH, D2O processor and 512 RAM.
exchangeable); 13C NMR (DMSO-d6) δ 14.6, 62.0, 108.6,
119.2, 122.2, 123.1, 124.8, 128.1, 128.6, 129.1, 133.9, Preparing for docking
140.2, 151.8, 153.2, 164.5, 166.2 ppm; MS, (relative
þ þ
abundance %): 449 (M  , 15.67 %), 451 (M  +2, 18.54%), The X-ray crystallographic structure of the Tyrosine Kinase
64 (100%); Anal. Calcd. for C19H14BrClN2O4 (449): C, Domain from EGFR TK complexed with erlotinib was
50.75; H, 3.14; N, 6.23. Found: C, 50.93; H, 3.34; N, 5.94. obtained from the Protein Data Bank (PDBID: 1M17).

Biological assay Validation of docking procedure

The two human cancer cell lines used in this study breast Docking of the co-crysatallized ligand should be carried out
carcinoma (MCF-7) and non-small cell lung cancer (A549) to study the scoring energy (s), root mean square deviation
were purchased from American Type Culture Collection. (RMSD), and amino acid interactions. The root mean
Cells were maintained in Dulbecco’s Modified Eagle’s square deviation (RMSD) which is measure of superposing
Medium (DMEM: Gibco, USA) supplemented with 10% was 0.50A° for the lead compound. Docking was performed
fetal bovine serum (Gibco), penicillin/streptomycin using London dG force and refinement of the results was
(Gibco).4′,6-Diamidino-2-phenylindole (DAPI) nuclear done using force field energy.
staining indicated that these cells were devoid of myco-
plasma contaminations. All incubations were done at 37 °C Preparing compounds for docking Preparing of synthe-
in a humidified incubator containing 5% CO2. Cells in sized compounds for docking was achieved through their

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


934 Med Chem Res (2017) 26:929–939

3D structure built by MOE. Certain procedures should be interactions and the hydrogen bond numbers and lengths
done before docking process including: 3D protonation of were summarized in (Table 2).
the structures, running conformational analysis using sys-
temic search, selecting the least energetic conformer, and
applying the same docking protocol used with ligand.
The previous measures were taken and docking for Results and discussion
the synthesized compounds was applied. Amino acid
Chemistry
Table 1 In vitro cytotoxic activity of reference drugs doxorubicin,
erlotinib and 4-(4-substituted-anilino)quinoline derivatives 6a–f The target 4-anlinoquinoline derivatives (6a–f) were syn-
thesized using the reaction sequence illustrated in
R2 Scheme 1. Accordingly, condensation of the bromo deri-
vatives (1a, b) with diethyl ethoxymethylenemalonate
yielded the corresponding enamine derivatives (3a, b) via
Gould–Jacobs reaction. Thermal cyclization of (3a, b) in
HN boiling diphenyl ether yielded the respective quinolone
R1 COOC2H5 derivatives (4a, b) which undergo chlorination by phos-
phorus oxychloride gave 4-chloroquinoline derivatives (5a,
b). Nucleophilic substitution of the active chlorine in (5a, b)
N with the appropriate aromatic amines gave the target 4-
6a-f anlinoquinoline derivatives (6a–f) in moderate yields
R (62–71%).
Compound no. IC50a (µM) Enamine derivatives (3a, b) were confirmed by IR
spectra which showed absorption band at 3486–3130 cm−1
R R1 R2 MCF-7 A549
revealed NH group presence. The 1H NMR spectra dis-
Doxorubicin – – – 2.07 1.14
played two doublet signals, each of one proton intensity at δ
Erlotinib – – – 0.02b 19.26c
8.50–8.54 ppm and 11.21–11.40 ppm with J value = 12.6
6a Br COOH CH3 5.51 7.31 Hz indicating the olefinic CH and NH respectively. These
6b Br COOH OCH3 10.84 11.26 characteristic two doublet signals disappeared in the qui-
6c Br COOH Cl 3.42 5.97 nolone derivatives (4a, b). The IR spectra of the quinoli-
6d COOH Br CH3 18.11 16.32 none derivatives (4a, b) showed sharp peak at 1636–1648
6e COOH Br OCH3 23.32 22.01 cm−1 corresponding to C=O which disappeared in the
6f COOHC Br Cl 14.53 12.32 chloroquinoline compounds (5a, b). Additionally, 13C
a
IC50: concentration of a drug that is required for 50% inhibition of NMR spectra of (4a, b) confirmed the presence of CH3 and
cell viability CH2 carbons at δ 14.2–14.3 ppm and at δ 60.6–60.9 ppm,
b
Data obtained from literature (Lv et al. 2010) also deshielded peak at δ 164.4 ppm of quinolone
c
Data obtained from literature (XQ and TK 2013) C=O which disappeared in the chloroquinoline derivatives

Table 2 Molecular modeling


Compound No. Number of H- Atoms of compound Amino acid residues Binding energy
data for compounds 6a, 6c, 6e
bonds forming H-bonds forming H-bonds(H-bond score(Kcal/mol)
and erlotinib during docking in
length in Ao)
the EGFR active sites
N1 Thr-766 (2.78 Ao) −11.37
Erlotinib 2 N3 Met-769 (2.70 Ao)
6a 2 O (acid) Thr-766 (2.55 Ao) −10.04
C=O (acid) Met-769 (2.44 Ao)
6e 3 C=O (ester) Thr-766 (2.94 Ao)
N (quinoline) Lys-721 (2.47 Ao) −10.78
C=O (acid) Lys-721 (2.19 Ao)
6c 3 O (acid) Thr-766 (2.38 Ao)
C=O (acid) Met-769 (2.82 Ao) −9.82
NH Asp-831 (1.73 Ao)

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Med Chem Res (2017) 26:929–939 935

R respectively confirmed presence of the OCH3 group of 4-


NH2 H COOC2H5 anilinophenyl ring. Finally, the mass spectra of (3a, b, 4a,
+ C C þ
C2H5O COOC2H5 b, 5a, b, and 6a–f) demonstrated molecular ion (M  ) peaks
1
R for all compounds as well as additional peaks with relative
1 a,b 2
abundance equal that of the molecular ion corresponding to
1a, R = Br, R1 = COOH a
1b, R = COOH, R1 = Br M+2 due to presence of bromine.

R COOC2H5
NH
COOC2H5 Biological results
3 a,b
1
R
b
All the target compounds (6a–f) were screened for their
in vitro anticancer activity against human breast cancer cell
O
line (MCF-7) and non-small cell lung cancer cell line
R1 COOC2H5
(A549) using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-
2H-tetrazolium bromide (MTT) assay (Omar et al. 2013).
N
The relationship between survival ratio and drug con-
R H
centration was plotted to obtain the survival curve of the
4a,b used cell lines (MCF-7 and A549). The response parameter
calculated was IC50 value (Table 1), which corresponds to
c the compound concentration causing 50% mortality in net
cells. The obtained data revealed that the target compounds
Cl
(6a–f) showed activities against MCF-7 and A549 cell lines
R1 COOC2H5
(IC50 = 3.42–23.32 and 5.97–22.01 µM respectively) in
N
comparison with doxorubicin (IC50 = 2.07 and 0.02 µM
R
respectively) and erlotinib (IC50 = 1.14 and 19.26 µM
5a,b respectively). For both cell lines, the 6-carboxy-8-bromo
analogs (6a, 6b and 6c) exhibited higher potency (IC50 =
d R2
5.51, 10.84, and 3.42 µM respectively for MCF-7 and IC50
= 7.31, 11.26, and 5.97 µM respectively for A549) rather
HN than 6-bromo-8-carboxy analogs (6d, 6e, and 6f) (IC50 =
R1 COOC2H5 18.11, 23.32, and 14.53 µM respectively for MCF-7 and
IC50 = 16.23, 22.01, and 12.32 µM respectively for A549).
N These results revealed that presence of carboxylic group at
6a-f
R position 6 of quinoline ring enhance the activity through
6a, R = Br, R1 = COOH, R2 =CH3; 6b, R = Br, R1 = COOH, R2 =
formation of additional hydrogen bonds with certain amino
OCH3
6c, R = Br, R1 = COOH, R2 = Cl; 6d, R = COOH, R1 = Br, R2 = acids in the active site of EGFR. On the other hand, para
CH3; substitution at aniline moiety with Cl as an electron with-
6e, R = COOH, R1 = Br, R2 = OCH3; 6f, R = COOH, R1= Br, R2 = Cl
drawing group was favored over CH3 or OCH3 substitution
Scheme 1 Synthesis of 4-(4-substituted-anilino)quinoline derivatives since (6c and 6f) were the most active compounds followed
6a–f by CH3 containing derivatives (6a and 6d) then the meth-
oxy derivatives (6b and 6e). Compound (6c) was the most
active derivative with IC50 = 3.42 and 5.97 µM against
(5a, b). The target compounds (6a–f) structure confirmed MCF-7 and A549 cell lines respectively that can be
by the presence of the additional aromatic moieties by explained by the presence of COOH group in position 6 of
presence of two doublet peaks at δ 6.87–7.21 ppm with J quinoline ring, in addition of para substitution of pheny-
value = 8.4–8.8 Hz of phenyl H-2,H-6 and at δ 7.07–7.53 lamino by Cl. Finally, all tested compounds 6a–f had higher
ppm with J value = 8.4–8.8 Hz of phenyl H-3,H-5 in the 1H activity against non-small cell lung cancer cell line (A549)
NMR spectra. Compounds (6a, d) had additional single (IC50 = 5.97–16.23 µM) than erlotinib (IC50 = 19.26 µM)
signal at δ 2.35, 2.78 ppm corresponding to CH3 group at 4- except 6e which was approximately equipotent (IC50 =
anilinophenyl ring in their 1H NMR spectra, while it 19.26 µM) that could be attributed to presence of halogen Br
appeared in 13C NMR spectra at δ 20.9 ppm. 1H NMR and in position 6 of quinoline ring instead of COOH group and
13
C NMR spectra of compounds (6b, e) showed presence of also 4-phenylamino substitution by methoxy group which
a single peak at δ 3.10, 3.80 ppm and at δ 48.8, 55.4 ppm caused decreased activity.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


936 Med Chem Res (2017) 26:929–939

Docking studies the docking reliability. We obtained the X-ray structure of


TK domain of epidermal growth factor receptor (EGFR TK)
A molecular modeling study has been performed to under- in complex with erlotinib which is the ligand from protein
stand the protein kinase inhibitor interactions and the struc- data bank (PDB ID: 1M17). Using MOE version 2008.10 the
tural features of the ATP active site of EGFR. Also, to ligand erlotinib was docked to the binding site of EGFR TK,
investigate the possible binding conformation for the newly and we selected the lowest energy score docking conformer
prepared 4-anlinoquinoline derivatives to ATP binding site of as the most probable binding conformation. In addition its
EGFR, this may give a suggestion about their proposed interaction mode revealed that two amino acids are involved
mechanism of action. First of all, it is necessary to validate in the interaction; Thr-766 and Met-769 (Fig. 2).

Fig. 2 Binding of erlotinib with EGFR a the proposed binding mode inside the active site of EGFR resulting from docking, the most important
amino acids are shown together with their respective numbers. b 2D interaction

Fig. 3 Binding of 6a with EGFR a the proposed binding mode inside the active site of EGFR resulting from docking, the most important amino
acids are shown together with their respective numbers. b 2D interaction

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Med Chem Res (2017) 26:929–939 937

We then docked the most in vitro cytotoxic active hydrogen bonding interactions while the score of compound
compounds (6-carboxy-8-bromo analogs, 6a and 6c) and 6c = −9.82 and formed three hydrogen bonding interac-
the least active compound (6-bromo-8-carboxy analog 6e). tions. Compound (6e) showed −10.78 Kcal/mol score with
The docking results including the energy score associated three hydrogen bonding interactions in comparison with
with intermolecular interactions obtained upon computa- erlotinib (I) which had energy score = −11.37 Kcal/mol
tional docking for the three compounds (6a, 6c, and 6e) and and formed two hydrogen bonding interactions. Compound
the hydrogen bonding interactions between the amino acid (6a) formed a hydrogen bond between carbonyl group of
residues and functional groups of the compounds are COOH group and Met 769 whiles the other hydrogen bond
summarized in Table 2. with Thr 766 through the remaining O of COOH. These two
From the docking results, both compounds (6a and 6c) hydrogen bonding interactions were observed similarly in
showed good binding interactions affinity; compound (6a) erlotinib (I). Compound (6c) formed the same two hydrogen
had a Glide score of −10.04 Kcal/mol and exhibited two bonding interactions with the same amino acid residues in

Fig. 4 Binding of 6c with EGFR a the proposed binding mode inside the active site of EGFR resulting from docking, the most important amino
acids are shown together with their respective numbers. b 2D interaction

Fig. 5 Binding of 6e with EGFR a the proposed binding mode inside the active site of EGFR resulting from docking, the most important amino
acids are shown together with their respective numbers. b 2D interaction

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


938 Med Chem Res (2017) 26:929–939

addition to a third one between NH and Asp 831. The least Afzal O, Kumar S, Haider MR, Ali MR, Kumar R, Jaggi M, Bawa S
(2015) A review on anticancer potential of bioactive heterocycle
in vitro cytotoxic active compound (6e) formed only one quinoline. Eur J Med Chem 97:871–910
hydrogen bond of 2 hydrogen bonding interactions of the Amin KM, Georgey HH, Awadallah FM (2011) EGFR tyrosine kinase
ligand (carbonyl group of the ester moiety with Thr 766) targeted compounds: synthesis, docking study, and in vitro anti-
and also two other hydrogen bonding interactions not pre- tumor activity of some new quinazoline and benzo[d]isothiazole
derivatives. Med Chem Res 20:1042–1053
sent in the ligand binding with Lys 721. These docking
Arora A, Scholar EM (2005) Role of tyrosine kinase inhibitors in
results suggest that compounds 6a and 6c (carboxylic group cancer therapy. J Pharmacol Exp Ther 315:971–979
in position 6 of quinoline moiety) may exhibit their cyto- Ekoue K, Yearick K, Iwaniuk DP, Natarajan JK, Alumasa J, De AC,
toxicity in a similar manner to erlotinib (Figs. 3–5). Roepe PD, Wolf C (2009) Synthesis and antimalarial activity of
new 4-amino-7-chloroquinolyl amides, sulfonamides, ureas and
thioureas. Bioorg Med Chem 17:270–283
El-Feky SAH, Abd El-Samii ZK, Osman NA, Lashine J, Kamel MA,
Conclusion Thabet HK (2015) Synthesis, molecular docking and anti-
inflammatory screening of novel quinoline incorporated pyr-
azole derivatives using the Pfitzinger reaction II. Bioorg Chem
A new series of 4-(4-substituted-anilino)quinoline deriva- 58:104–116
tives (6a–f) was designed and synthesized for evaluation as Ferlin MG, Chiarelotto G, Dall’Acqua S, Maciocco E, Mascia MP,
anticancer against two human cancer cell lines; breast car- Pisu MG, Biggio G (2005) Novel anellated pyrazoloquinolin-3-
ones: synthesis and in vitro BZR activity. Bioorg Med Chem
cinoma (MCF-7) and non-small cell lung cancer (A549).
13:3531–3541
Structure-activity data and molecular modeling studies Gouda AM, Abdelazeem AH, Arafa EA, Abdellatif KRA (2014)
showed that (i) the 6-carboxy-8-bromo analogs (6a, 6b, and Design, synthesis and biological evaluation of novel triaryl (Z)-
6c) exhibited higher potency than 6-bromo-8-carboxy ana- olefins as tamoxifen analogues. Bioorg Chem 53:1–7
Hamaguchi W, Masuda N, Miyamoto S, Shiina Y, Kikuchi S, Mihara
logs (6d, 6e, and 6f) which mains presence of carboxylic
T, Moriguchi H, Fushiki Y, Murakami Y, Amano Y, Honbou K,
group in position 6 of quinoline ring is favored over its Hattori K (2015) Synthesis, SAR study, and biological evaluation
presence in position 8, (ii) substitution at para position of of novel quinoline derivatives as phosphodiesterase 10A inhibi-
the aniline moiety with Cl as electron with drawing group is tors with reduced CYP3A4 inhibition. Bioorg Med Chem
23:297–313
favored over substitution with CH3 (electron donating
Hicks JW, Van HF, Wilson AA, Houle S, Vasdec N (2010) Radi-
group) and over substitution with OCH3 (electron donating olabeled small molecule protein kinase inhibitors for imaging
group) since 6c > 6a > 6b and 6f > 6d > 6e in potency, (iii) with PET or SPECT. Molecules 15:8260–8278
all tested compounds (6a–f) had higher activity against non- Lv P, Zhou C, Chen J, Liu P, Wang K, Mao W, Li H, Yang Y, Xiong
J, Zhu H (2010) Design, synthesis and biological evaluation of
small cell lung cancer cell line (A549) than erlotinib except
thiazolidinone derivatives as potential EGFR and HER-2 kinase
(6e) which was approximately equipotent, (iv) molecular inhibitors. Bioorg Med Chem 18:314–319
docking for the most in vitro cytotoxic active compounds Ma X, Zhou W, Brun R (2009) Synthesis, in vitro antitrypanosomal
(6a and 6c) revealed that these compounds may exhibit their and antibacterial activity of phenoxy, phenylthio or benzyloxy
substituted quinolones. Bioorg Med Chem Lett 19:986–989
cytotoxicity in a similar manner to erlotinib and (v) the
Majerz K, Musiol R, Skórska A, Tabak D, Mazur P, Oleksyn BJ,
previous promising results could help in design of new Polanski J (2011) X-ray and molecular modelling in fragment-
potent quinoline derivatives targeting cancer chemotherapy. based design of three small quinoline scaffolds for HIV integrase
inhibitors. Bioorg Med Chem 19:1606–1612
Medapi B, Renuka J, Saxena S, Sridevi JP, Medishetti R, Kulkarni P,
Compliance with Ethical Standards Yogeeswari P, Sriram D (2015) Design and synthesis of novel
quinoline-aminopiperidine hybrid analogues as Mycobacterium
Conflict of interest The authors declare that they have no com- tuberculosis DNA gyraseB inhibitors. Bioorg Med Chem
peting interest. 23:2062–2078
Omar HA, Arafa SA, Salama SA, Arab HH, Wu CH, Weng JR (2013)
OSU-A9 inhibits angiogenesis in human umbilical vein endo-
thelial cells via disrupting Akt-NF-κB and MAPK signaling
References pathways. Toxicol Appl Pharm 272:616–624
Patel RV, Park SW (2014) Access to a new class of biologically active
Abdellatif KRA, Abdelall EKA, Abdelgawad MA, Ahmed RR, Bakr quinoline based 1,2,4-triazoles. Eur J Med Chem 71:24–30
RB (2014a) Synthesis, docking study and antitumor evaluation of Rano TA, Sieber E, Pelton PD, Yang M, Dermarest KT, Kuo G (2009)
certain newly synthesized pyrazolo[3,4-d]pyrimidine derivatives. Design and synthesis of potent inhibitors of cholesteryl ester
Org Chem Ind J 10:157–167 transfer protein (CETP) exploiting a 1,2,3,4-tetrahydroquinoline
Abdellatif KRA, Abdelall EKA, Abdelgawad MA, Ahmed RR, Bakr platform. Bioorg Med Chem Lett 19:2456–2460
RB (2014b) Synthesis and anticancer activity of some new pyr- Roma G, Grossi G, Di Braccio M, Piras D, Ballabeni V, Tongnolini
azolo[3,4-d]pyrimidin-4-one derivatives. Molecules M, Bertoni S, Barocelli E (2008) 1,8-naphthyridines VII. New
19:3297–3309 substituted 5-amino[1,2,4]triazolo[4,3-a][1,8]naphthyridine-6-
Abdellatif KRA, Belal A, Omar HA (2013) Design, synthesis and carboxamides and their isosteric analogues exhibiting notable
biological evaluation of novel triaryl (Z)-olefins as tamoxifen anti-inflammatory and/or analgesic activities, but no acute gas-
analogues. Bioorg Med Chem Lett 23:4960–4963 trolesivity. Eur J Med Chem 43:1665–1680

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Med Chem Res (2017) 26:929–939 939

Rossiter S, Péron J, Whitfield PJ, Jones K (2005) Synthesis and 1,3,4-oxadiazole-2(3H)-thione quinolone derivatives as novel
anthelmintic properties of arylquinolines with activity against anticancer agent. Eur J Med Chem 60:23–28
drug-resistant nematodes. Bioorg Med Chem Lett 15:4806–4808 XQ L, TK Q (2013) Radiosensitizing effect of erlotinib on human
Solomon VR, Lee H (2011) Quinoline as a privileged scaffold in lung adenocarcinoma cell line A549. Chinese J Oncol
cancer drug discovery. Curr Med Chem 18:1488–1508 35:819–823
Sun J, Zhu H, Yang Z, Zhu H (2013) Synthesis, molecular modeling
and biological evaluation of 2-aminomethyl-5-(quinolin-2-yl)-

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:

1. use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
2. use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
3. falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
4. use bots or other automated methods to access the content or redirect messages
5. override any security feature or exclusionary protocol; or
6. share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at

onlineservice@springernature.com

You might also like