Open Access Article
Magda M. F. Ismail
a,
Taghreed Z. Shawera,
Rabab S. Ibrahima,
Mostafa S. Abusaifb,
Mona M. Kamalc,
Rasha M. Allamd and
Yousry A. Ammarb
aDepartment of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt. E-mail: m.elalfy101@gmail.com; magdaismail@azhar.edu.eg
bDepartment of Chemistry, Faculty of Science, Al-Azhar University, Cairo, 11754, Egypt
cDepartment of Pharmacology, Faculty of Pharmacy (Girls), 11754 Al-Azhar University, Cairo, Egypt
dDepartment of Pharmacology, Medical and Clinical Research Institute, National Research Centre, 12622, Dokki, Cairo, Egypt
First published on 31st October 2023
Vascular endothelial growth factor receptor-2 is a vital target for therapeutic mediation in various types of cancer. This study was aimed at exploring the cytotoxic activity of seventeen novel quinoxaline-3-propanamides against colon cancer (HCT-116) and breast cancer (MCF-7) using MTT assay. Results revealed that compounds 8, 9, and 14 elicited higher cytotoxicity than the reference drugs, doxorubicin (DOX) and sorafenib. Interestingly, they are more selective for HCT-116 (SI 11.98–19.97) and MCF-7 (SI 12.44–23.87) compared to DOX (SI HCT-116 0.72 and MCF-7 0.9). These compounds effectively reduced vascular endothelial growth factor receptor-2; among them, compound 14 displayed similar VEGFR-2 inhibitory activity to sorafenib (IC50 0.076 M). The ability of 14 to inhibit angiogenesis was demonstrated by a reduction in VEGF-A level compared to control. Furthermore, it induced a significant increase in the percentage of cells at pre-G1 phase by almost 1.38 folds (which could be indicative of apoptosis) and an increase in G2/M by 3.59 folds compared to the control experiment. A flow cytometry assay revealed that compound 14 triggered apoptosis via the programmed cell death and necrotic pathways. Besides, it caused a remarkable increase in apoptotic markers, i.e., caspase-3 p53 and BAX. When compared to the control, significant increase in the expression levels of caspase-3 from 47.88 to 423.10 and p53 from 22.19 to 345.83 pg per ml in MCF-7 cells. As well, it increased the proapoptotic protein BAX by 4.3 times while lowering the antiapoptotic marker BCL2 by 0.45 fold. Docking studies further supported the mechanism, where compound 14 showed good binding to the essential amino acids in the active site of VEGFR-2. Pharmacokinetic properties showed the privilege of these hits over sunitinib: they are not substrates of P-gp protein; this suggests that they have less chance to efflux out of the cell, committing maximum effect; and in addition, they do not allow permeation to the BBB.
Quinoxaline derivatives are a widespread class of heterocycles receiving the most attention, especially in the field of chemotherapy.13–16 Many drugs incorporating the quinoxaline scaffold have achieved promising results and have been submitted to clinical trials for anticancer therapeutic purposes.17,18
The therapeutic applicability of current VEGFR inhibitors, however, is constrained to some extent by their low clinical efficacy and probable toxicity. Based on the aforementioned findings (cf. Fig. 1) and as a continuation of our prior work in the field of the design and synthesis of new anticancer medicines,19–26 a new series of 6-chloroquinoxaline-3-propanamides have been designed as sunitinib's mimetic to optimize pharmacodynamics properties and to overcome certain pharmacokinetic problems demonstrated on sunitinib clinical use such as, BBB permeation and being P-gp substrate.
Herein, the hetero-aromatic system was designed to be 6-chloroquinoxalin-2-one as an isostere for 5-fluoroindoline-2-one, and the linker group was an aliphatic chain (–CH2CH2–) as a chain contraction to replace 2,4-dimethyl-1H-pyrrole of sunitinib. The pharmacophore moiety was kept in all the designed compounds as an amide group. The terminal hydrophobic moiety was an aryl moiety with a variation of substituents instead of an aliphatic amino side chain of sunitinib. These modifications were followed to optimize both the pharmacodynamics and pharmacokinetic criteria of sunitinib, (Fig. 1).
![]() | ||
| Scheme 1 Reagents and conditions, (i) acetic acid/EtOH, stirr, r.t. 15 min (ii) CHsOH, dps conc. H2SO4, reflux, 3 h, (iii) aromatic amines, EtOH/DMF, reflux, 8–16 h. | ||
Additionally, aminolysis28 of ethyl propanoate ester 4 with substituted anilines at positions 2, 3, or 4 was carried out to yield the corresponding quinoxaline-3-propanamides 5–17. A single molecule of methanol was eliminated through reflux in a catalytic amount of DMF in ethanol solvent over the course of eight to sixteen hours, successfully completing the reaction despite aniline's very weak nucleophilicity towards the carbonyl carbon of the ester group, (Scheme 1).
The spectroscopic and micro-analytical data from all quinoxaline-3-propanamides 5–17 were used to elucidate them. All of our series are characterized by the presence of two triplets in the 1H NMR spectra that indicate linker protons (–CH2–CH2–CO) at δ (ppm): 1.63–1.75 for CH2 and 2.25–2.38 for CH2CO protons. Besides, deshielded signals for three quinox-Hs and four aryl-Hs are appeared at their appropriate aromatic region. It is important to note several distinctive signals in the 1H NMR spectra of our series; for example, the 4-CH3 protons of compound 11 appeared as a singlet at around δ 2.40 ppm, while the 4-OCH3 protons of compound 14 appeared at about δ 3.90 ppm. Compound 15's 1H NMR spectrum showed a singlet signal at about δ 2.67 ppm that was attributed to COCH3 protons. In the 1H NMR spectrum of the ethyl ester 17, a triplet–quartet pattern was seen at δ 1.34 and 4.36 ppm respectively.
Hydrazinolysis of the ester 4 with hydrazine hydrate using absolute ethanol produced a good yield of the corresponding acid hydrazide derivative, 18.29 Nucleophilic substitution reaction of compound 18 with 2,4-dichlorobenzoyl chloride was achieved by heating under reflux condition in ethanol with DMF drops30 to furnish the corresponding 2,4-dichlorobenzoyl hydrazide, 19 (Scheme 2). Furthermore, synthesis of the target compounds 20 and 21 was achieved by treatment of the intermediate 18 with the appropriate acid anhydrides namely; succinic anhydride or phthalic anhydride respectively via ring opening amidation reaction in ethanol with catalytic amount of DMF.31 Compound 20 was verified based on spectral data; its IR spectrum displayed extra bands at 3360 and 1706 cm−1 contributed to OH and C
O of carboxyl group respectively. 1H NMR spectrum displayed extra two triplets at δ 2.38 and 2.59 ppm contributed to CH2CH2 protons of succinic acid moiety. As mentioned in literature,32 reaction of the starting material 18 with phthalic anhydride is a solvent dependent. Thus, when the reaction was carried out in ethanol; the product was formulated as carboxylic acid derivative 21. While performing the reaction in glacial acetic acid as a solvent, the corresponding isoindoline derivative 22 was obtained via ring closure due to condensation reaction. The isoindoline 22 was further confirmed by refluxing compound 21 in glacial acetic for 2 h to afford one and the same product (22). The structures of these compounds were consistent with spectral analysis. IR of compound 21 showed stretching bands around 3339 and 1705 cm−1 pointing to hydroxyl and carbonyl of COOH respectively. 1H NMR spectrum showed a characteristic singlet at δ 10.80 ppm attributed to carboxylic proton; also 4 extra signals (4 Ar–H) at the range from δ 7.44 to 8.74 ppm were observed. Mass spectrum exhibited a peak at m/z 414 referred to molecular ion peak (M+). Concerning IR spectrum of compound 22, lack of broad band of OH and NH is observed due to removal of water molecule. In addition, the frequency of carbonyl band is increased due to ring closure, where 1H NMR spectrum displayed only two signals at 12.22 and 12.54 ppm for two NH protons with lack of COOH and one NH signals ensures the cyclization (Scheme 2).
| Compd. no. | HCT-116 | MCF-7 | Compd. no. | HCT-116 | MCF-7 |
|---|---|---|---|---|---|
| a IC50 (μM): 1–10 (very strong). 11–20 (strong). 21–50 (moderate). 51–100 (weak) and above 100 (non-cytotoxic).b DOX: Doxorubicin. | |||||
| DOX | 9.27 ± 0.3 | 7.43 ± 0.2 | 12 | 28.35 ± 2.2 | 24.56 ± 1.9 |
| Sorafenib | 18.60 ± 1.9 | 16.0 ± 3.6 | 13 | 22.19 ± 1.9 | 18.88 ± 1.4 |
| Sunitinib | 3.42 ± 0.5 | 4.77 ± 0.2 | 14 | 5.81 ± 0.5 | 4.61 ± 1.1 |
| 5 | 45.52 ± 2.7 | 39.03 ± 2.4 | 15 | 84.46 ± 3.9 | 76.73 ± 3.8 |
| 6 | 62.21 ± 3.3 | 54.73 ± 3.2 | 16 | 75.06 ± 3.6 | 67.23 ± 3.5 |
| 7 | 93.14 ± 4.6 | 80.39 ± 4.1 | 17 | 36.02 ± 2.4 | 31.17 ± 2.2 |
| 8 | 4.72 ± 0.8 | 3.95 ± 0.2 | 19 | 6.18 ± 2.1 | 5.11 ± 0.8 |
| 9 | 7.19 ± 0.7 | 6.92 ± 0.5 | 20 | 26.41 ± 3.8 | 10.17 ± 3.3 |
| 10 | 19.41 ± 1.5 | 11.93 ± 0.9 | 21 | 48.17 ± 2.7 | 41.47 ± 2.5 |
| 11 | 61.35 ± 3.4 | 53.95 ± 3.1 | 22 | 37.2 ± 2.3 | 25.92 ± 1.9 |
| Compd no. | WI-38 | HCT-116 | SI | MCE-7 | SI | VEGFR-2 |
|---|---|---|---|---|---|---|
| 8 | 94.27 ± 3.5 | 4.72 ± 0.8 | 19.97 | 3.95 ± 0.2 | 23.87 | 0.109 |
| 9 | 86.11 ± 4.1 | 7.19 ± 0.7 | 11.98 | 6.92 ± 0.5 | 12.44 | 0.131 |
| 14 | 88.72 ± 3.9 | 5.81 ± 0.5 | 15.27 | 4.61 ± 1.1 | 19.25 | 0.076 |
| 19 | 71.60 ± 2.8 | 6.18 ± 2.1 | 11.58 | 5.11 ± 0.8 | 14.01 | — |
| 20 | 93.04 ± 2.2 | 26.41 ± 3.8 | 3.52 | 10.17 ± 3.3 | 9.15 | — |
| DOX | 6.72 ± 0.5 | 9.27 ± 0.3 | 0.72 | 7.43 ± 0.2 | 0.90 | — |
| Sunitinib | — | 3.42 ± 0.5 | — | 4.77 ± 0.2 | — | 0.139 (ref. 35) |
| Sorafenib | — | 18.60 ± 1.9 | — | 16.0 ± 3.6 | — | 0.076 |
In regard to positional isomers, the activity's order was 13 (2-CH3) > 12 (3-CH3) > 11 (4-CH3). The high potency is correlated with electron-releasing by +I of CH3 at p-2 (the preferred position), thus the electron-releasing is a beneficial criteria for cytotoxicity. Conversely, it appeared from the IC50s of compounds 8 (4-OH) and 9 (2-OH) that 8 was slightly more active; this may be attributed to the fact that 4-OH affects the activity via both +I and +M effects rather than 2-OH, which shows only +I effects.
Another polar but highly electron-withdrawing compound is compound 10 (4-NO2), which displayed strong cytotoxicity (IC50 19.41, 11.93 μM), towards HCT116 and MCF-7, respectively. Additionally, moderate activities appeared for compounds 5 (4-H) and 17 (4-COOEt), while both the analogues 15 (4-COCH3) and 16 (4-COOH) bearing strong electron-withdrawing groups are weak cytotoxic agents. Regarding compound 19, very strong cytotoxicity results (IC50 6.18, 5.11 μM) against HCT116 and MCF-7, respectively was noticed. Finally, the cyclized phthalimide derivative 22 exhibited better antitumor activity than the open-chain one, 21 (Table 1).
| Sample | Total | Early apoptosis | Late apoptosis | Necrosis |
|---|---|---|---|---|
| 14/MCF-7 | 22.03 | 2.05 | 6.25 | 12.73 |
| Control/MCF-7 | 2.17 | 0.15 | 0.73 | 1.29 |
![]() | ||
| Fig. 6 (a) Effect of control on apoptosis of MCF-7 cells. (b) Effect of 14 on apoptosis of MCF-7 cells. | ||
The quadrants in the cytograms represent the following: Necrotic cells (higher left quadrant of the cytogram); late apoptotic cells (higher right quadrant of the cytogram); non-apoptotic and non-necrotic cells (living cells) (lower left quadrant of the cytogram); early apoptotic cells (lower right quadrant of the cytogram).
| Sample | Caspase 3 (pg ml−1) | Fld | p53 (pg ml−1) | Fld | BAX fold change | BCL2 fold change |
|---|---|---|---|---|---|---|
| 14 | 423.10 ± 11.24 | 8.836 | 345.83 ± 7.44 | 15.585 | 4.336 | 0.448 |
| Sunitinib | 384.53 ± 8.40 | 8.031 | 278.34 ± 5.27 | 12.543 | — | — |
| Staurosporine | — | — | — | — | 7.5517 | 0.3248 |
| Control | 47.88 ± 13.15 | 1 | 22.19 ± 8.39 | 1 | 1 | 1 |
Here, using the online version of SwissADME, a computational assessment of the synthesized compounds was carried out to assess the physicochemical and ADME properties.47 Regarding Lipinski's rule for oral medications, which indicates that a molecule is more likely to be absorbed or permeated if its molecular weight is less than 500 g mol−1, its M
log
P value is lower than 5, and it has at least 5 H-donor and 10 H-acceptor atoms.48 While Veber's rule49 specifies polar surface area (PSA) 140 and rotatable bond count 10 as drug-likeness restrictions, respectively.50 It was discovered that all of the synthesized compounds have Lipinski zero violations in their physicochemical parameters and every chemical has rotatable bonds between 5 and 8 that indicate molecular flexibility for its bio target (Table 5). In addition, all hits pass muster under the Veber guidelines used for screening.
| Cpd. no. | HBD | HBA | M log P |
MWt | No. of Rot. bonds | Lipinski's violations | Veber's violations |
|---|---|---|---|---|---|---|---|
| 5 | 2 | 3 | 2.39 | 327.76 | 5 | 0 | 0 |
| 6 | 2 | 3 | 2.89 | 362.21 | 5 | 0 | 0 |
| 7 | 2 | 4 | 2.78 | 345.76 | 5 | 0 | 0 |
| 8 | 3 | 4 | 1.85 | 343.76 | 5 | 0 | 0 |
| 9 | 3 | 4 | 1.85 | 343.76 | 5 | 0 | 0 |
| 10 | 2 | 5 | 1.40 | 372.76 | 6 | 0 | 0 |
| 11 | 2 | 3 | 2.63 | 341.79 | 5 | 0 | 0 |
| 12 | 2 | 3 | 2.63 | 341.79 | 5 | 0 | 0 |
| 13 | 2 | 3 | 2.63 | 341.79 | 5 | 0 | 0 |
| 14 | 2 | 4 | 2.08 | 357.79 | 6 | 0 | 0 |
| 15 | 2 | 4 | 1.97 | 369.80 | 6 | 0 | 0 |
| 16 | 3 | 5 | 2.02 | 371.77 | 6 | 0 | 0 |
| 17 | 2 | 5 | 2.47 | 399.83 | 8 | 0 | 0 |
| 19 | 3 | 4 | 3.15 | 439.68 | 7 | 0 | 0 |
| 20 | 4 | 6 | 0.57 | 366.76 | 9 | 0 | 1 |
| 21 | 4 | 6 | 1.83 | 414.80 | 8 | 0 | 1 |
| 22 | 2 | 5 | 1.93 | 396.78 | 5 | 0 | 0 |
| Sorafenib | 3 | 7 | 2.91 | 464.82 | 9 | 0 | 0 |
| Sunitinib | 3 | 4 | 2.06 | 398.47 | 8 | 0 | 0 |
The topological polar surface area (TPSA) is known to be a reliable indication of drug penetration through the blood–brain barrier (TPSA less than 60 Å2) and intestinal drug absorption (TPSA less than 140 Å2). All compounds exhibit computational TPSA values in the appropriate range for permeating cell membranes so they obey Veber's rule except compounds 20 and 21 (TPSA 141 Å2). Moreover, absorption (% ABS) was calculated using the formula% ABS = 109 − (0.345 × TPSA), and it was discovered that the calculated % ABS of all these hits ranged between 60.26% and 83.17%, indicating that these synthetic derivatives may have the necessary cell membrane permeability and bioavailability (Table 6).
| Comp. no. | TPSA | ABS% |
|---|---|---|
| 5 | 74.85 | 83.17 |
| 6 | 74.85 | 83.17 |
| 7 | 74.85 | 83.17 |
| 8 | 95.08 | 76.19 |
| 9 | 95.08 | 76.19 |
| 10 | 120.67 | 67.36 |
| 11 | 74.85 | 83.17 |
| 12 | 74.85 | 83.17 |
| 13 | 74.85 | 83.17 |
| 14 | 84.08 | 79.99 |
| 15 | 91.92 | 77.28 |
| 16 | 112.15 | 70.30 |
| 17 | 101.15 | 74.10 |
| 19 | 103.95 | 73.13 |
| 20 | 141.25 | 60.26 |
| 21 | 141.25 | 60.26 |
| 22 | 112.23 | 70.28 |
| Sorafenib | 92.35 | 77.13 |
| Sunitinib | 77.23 | 108.92 |
The synthesized compounds' pharmacokinetic and medicinal chemistry parameters (Table 7) made it clear that all of the derivatives had high gastrointestinal absorption and that the majority of them did not cross the blood–brain barrier, guaranteeing that these systemically targeted molecules would have few to no CNS side effects. Analyzing the P-glycoprotein (P-gp) non-substrate candidature during preclinical analysis trials was another crucial factor. P-gp performs the function of an efflux transporter, expelling medicines, other substances, and its substrate from the cell. This sounds like an explanation for why anticancer chemotherapeutic drugs like Imatinib, Lonafarnib, and Taxanes are ineffective. The fact that not all hits are P-gp protein substrates (Table 7) suggests that these hits have a reduced likelihood of effluxing out of the cell and committing maximal effect.
| Compd no. | GI absorption | BBB permeation | P-gp substrate | Bioavailability score | PAINS alerts | Synthetic accessibility |
|---|---|---|---|---|---|---|
| 5 | High | Yes | No | 0.55 | 0 | 2.58 |
| 6 | High | Yes | No | 0.55 | 0 | 2.60 |
| 7 | High | Yes | No | 0.55 | 0 | 2.59 |
| 8 | High | No | No | 0.55 | 0 | 2.57 |
| 9 | High | No | No | 0.55 | 0 | 2.61 |
| 10 | High | No | No | 0.55 | 0 | 2.68 |
| 11 | High | Yes | No | 0.55 | 0 | 2.63 |
| 12 | High | Yes | No | 0.55 | 0 | 2.65 |
| 13 | High | Yes | No | 0.55 | 0 | 2.66 |
| 14 | High | No | No | 0.55 | 0 | 2.66 |
| 15 | High | No | No | 0.55 | 0 | 2.69 |
| 16 | High | No | No | 0.56 | 0 | 2.60 |
| 17 | High | No | No | 0.55 | 0 | 2.85 |
| 19 | High | No | No | 0.55 | 0 | 2.96 |
| 20 | High | No | Yes | 0.56 | 0 | 2.91 |
| 21 | High | No | Yes | 0.56 | 0 | 2.98 |
| 22 | High | No | No | 0.55 | 0 | 2.83 |
| Sorafenib | Low | No | No | 0.55 | 0 | 2.87 |
| Sunitinib | High | Yes | Yes | 0.55 | 0 | 3.58 |
The amount of drug present in the plasma is measured by bioavailability, which is regarded as the most important factor influencing absorption. It's interesting to note that all of the synthetic compounds have high bioavailability ratings on par with sorafenib and sunitinib.
Chemical substances known as pan-assay interference chemicals (PAINS) frequently produce false-positive findings in high-throughput screening. Instead of directly affecting one intended target, PAINS typically respond non-specifically with many biological targets. Checking any PAINS alert of the recently created derivatives is crucial. SwissADME did a PAIN that showed no alarms for any of the hits. The SwissADME Synthetic Accessibility (SA) Score is based mainly on the supposition that the frequency of molecular fragments in ‘really’ attainable molecules correlates with the ease of synthesis; the score is normalized to range from 1 (very easy) to 10 (very difficult). SA scores of all the analogues were found to be between 2.57 and 2.85, indicating that they can be easily synthesized on a large scale.
The in silico ADME prediction analysis results showed that the synthesized compounds exhibit the computational assessment and are therefore viewed as a pharmacologically active framework that should be taken into consideration when moving forward with potential hits.
Using MOE 2014 software, it is possible to investigate the binding interaction of the target compounds using the known crystal structure of VEGFR-2 (PDB ID: 4ASD).52 Reference molecules were the co-crystallized ligands sorafenib and sunitinib. The results of the docking studies demonstrated that our hit 14 had a high affinity for VEGFR-2 in comparison to the reference molecules. Sorafenib, a co-crystallized ligand, was re-docked against VEGFR-2 to verify the docking techniques. The applied docking methodology was validated by the resulting RMSD of 0.88 (Fig. 8).
![]() | ||
| Fig. 8 The proposed 2D (left) and 3D (right) binding interaction of co-crystalized sorafenib with 4ASD. | ||
Sorafenib's docking score was −10.74 kcal mol−1, and the urea moiety's three hydrogen bonds with the critical amino acids Glu885, Asp1046, and Cys1045 allowed it to bind to the receptor. The hinge region, where the pyridine moiety established a hydrogen link with Cys919 and an arene–H bond with Leu840, was also occupied by the N-methylpicolinamide moiety. Additionally, Leu840, Val916, Leu889, Phe1047, and Leu1035 all showed five hydrophobic contacts (Fig. 8).
Sunitinib inhibits the ATP pocket by an H-bond acceptor/donor interaction between two HB of the indolin-2-one core and Cys919 and Glu917 in the hinge region, together with the pyrrole group as a linker connected to Gly922 via arene-H. Docking experiments showed that sunitinib had a binding energy of −5.76 kcal mol−1. Leu840, Phe1047, Leu1035, Gly922, Val916, and Phe918 were among the hydrophobic interactions that the sunitinib tail was involved in (Fig. 9).
![]() | ||
| Fig. 9 The proposed 2D (left) and 3D (right) binding interaction of the parent sunitinib with 4ASD. | ||
Our hit 14 had an incredibly low free binding energy of −11.58 kcal mol−1 when compared to sorafenib and sunitinib. It showed 2HBA between the quinox-CO, Phe918, and Cys919 in the hinge region. Quinox-NH was also shared by 1HBD with Glu917. On the other hand, 1HBD also provided CH2 of the linker to the critical amino acid Cys919. Unexpectedly, the gate area's Asp1046 bound to the chloro functionality by HBD, therefore it is supposed that 14 may act as type II inhibitor of VEGFR-2. Additionally, several hydrophobic interactions involving the methoxyl group, terminal phenyl, and aliphatic linker helped the compound attach to VEGFR-2 (Fig. 10). These interactions included Gly922, Asn923, Val916, Phe1047, Le840, and Leu1035. Overlay docking alignment was also carried out between sorafenib (yellow), sunitinib (blue), and 14 (green) in the active site of VEGFR-2 to verify the target chemical 14's mode of action as a VEGFR-2 inhibitor (Fig. 11).
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.68 (t, 2H, CH2x, J = 7.6 Hz), 2.34 (t, 2H, CH2y, J = 7.6 Hz), 6.87 (t, 1H, Ar–H4), 7.07 (t, 2H, Ar–H3,5, J = 8 Hz), 7.41 (d, 2H, Ar–H2,6, J = 8 Hz), 7.60 (d, 1H, quinoxaline-H7, J = 8 Hz), 8.23 (s, 1H, quinoxaline-H5), 8.33 (d, 1H, quinoxaline-H8, J = 8 Hz), 10.31, 12.59 (2s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.34 (CH2), 31.50 (CH2), 119.12, 120.34, 120.50, 120.57, 125.45, 125.75, 126.12, 126.19, 126.45, 128.34, 129.50, 130.57, 131.12 (13 Ar–C), 167.12, 170.34 (2C
O). MS m/z: 327.12 (21.53%, M+), 329.14 (7.16%, M+2), 76.09 (100). Anal. calcd for: C17H14ClN3O2 (327.76): C, 62.30; H, 4.31; N, 12.82%, found: C, 62.73; H, 4.67; N, 12.40%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.66 (t, 2H, CH2x, J = 8 Hz), 2.33 (t, 2H, CH2y, J = 8 Hz), 7.14 (d, 1H, quinoxaline-H7, J = 7.6 Hz), 7.64 (d, 2H, Ar–H3,5, J = 8 Hz), 7.83 (d, 2H, Ar–H2,6, J = 8 Hz), 7.96 (s, 1H, quinoxaline-H5), 8.02 (d, 1H, quinoxaline-H8, J = 7.6 Hz), 11.51, 11.64 (2s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.03 (CH2), 30.81 (CH2), 121.68, 128.62, 128.69, 129.29, 129.34, 134.35, 134.36, 135.62, 136.28, 137.69, 145.29, 147.27, 148.21 (13 Ar–C), 164.09, 165.69 (2C
O). MS m/z: 361.53 (62.18%, M+), 363.20 (20.46%, M+2), 365.14 (8.06%, M+4), 74.03 (100). Anal. calcd for: C17H13Cl2N3O2 (362.21): C, 56.37; H, 3.62; N, 11.60%, found: C, 56.81; H, 3.93; N, 11.15%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.72 (t, 2H, CH2x, J = 8 Hz), 2.32 (t, 2H, CH2y, J = 8 Hz), 6.85 (d, 1H, quinoxaline-H7, J = 8.4 Hz), 6.99 (d, 2H, Ar–H2,6, J = 8 Hz), 7.40 (t, 2H, Ar–H3,5, J = 8 Hz), 7.60 (s, 1H, quinoxaline-H5), 7.80 (d, 1H, quinoxaline-H8, J = 8.4 Hz), 11.17, 11.32 (2 s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.46 (CH2), 31.28 (CH2), 151.57, 115.68, 116.70, 116.78, 118.78, 121.02, 121.78, 122.02, 122.25, 123.27, 131.18, 136.21, 143.32 (13 Ar–C), 166.10, 170.13 (2C
O). MS m/z: 345.04 (18.43%, M+), 347.11 (5.96%, M+2), 77.14 (100). Anal. calcd for: C17H13ClFN3O2 (345.46): C, 59.05; H, 3.79; N, 12.15%, found: C, 59.55; H, 3.23; N, 12.62%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.69 (t, 2H, CH2x, J = 7.2 Hz), 2.34 (t, 2H, CH2y, J = 7.2 Hz), 6.74 (d, 2H, Ar–H3,5, J = 8 Hz), 7.00 (d, 1H, quinoxaline-H7, J = 7.6 Hz), 7.37 (s, 1H, quinoxaline-H5), 7.50 (d, 2H, Ar–H2,6, J = 8 Hz), 7.78 (d, 1H, quinoxaline-H8, J = 7.6 Hz), 9.83, 12.37 and 12.48 (3s, OH, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.87 (CH2), 30.69 (CH2), 126.52, 127.33, 128.42, 129.87, 130.34, 132.37, 133.50, 134.24, 137.28, 137.29, 137.30, 140.71, 150.37 (13 Ar–C), 164.32, 168.33 (2C
O). MS m/z: 343.19 (27.20%, M+), 345.09 (9.56%, M+2), 71.93 (100). Anal. calcd for: C17H14ClN3O3 (343.76): C, 59.40; H, 4.10; N, 12.22%, found: C, 59.82; H, 4.43; N, 12.63%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.66 (t, 2H, CH2x, J = 7.6 Hz), 2.35 (t, 2H, CH2y, J = 7.6 Hz), 6.93 (t, 1H, Ar–H5, J = 8 Hz), 7.05 (d, 1H, Ar–H3, J = 8 Hz), 7.14 (t, 1H, Ar–H4, J = 8 Hz), 7.43 (d, 1H, quinoxaline-H7, J = 8 Hz), 7.63 (s, 1H, quinoxaline-H5), 7.64 (d, 1H, Ar–H6, J = 8 Hz), 7.83 (d, 1H, quinoxaline-H8, J = 8 Hz), 9.85, 11.09 and 11.12 (3 s, OH, 2NH D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.48 (CH2), 30.32 (CH2), 120.12, 126.32, 128.20, 129.40, 130.47, 134.60, 134.77, 135.47, 135.60, 135.78, 137.37, 137.38, 138.90 (13 Ar–C), 165.47, 170.50 (2C
O). MS m/z: 343.23 (31.89%, M+), 345.14 (10.03%, M+2), 89.1 (100). Anal. calcd for: C17H14ClN3O3 (343.76): C, 59.40; H, 4.10; N, 12.22%, found: C, 59.67; H, 4.38; N, 12.42%.
O), 1508, 1369 (NO2); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.75 (t, 2H, CH2x, J = 6.4 Hz), 2.35 (t, 2H, CH2y, J = 6.4 Hz), 7.39 (d, 1H, quinoxaline-H7, J = 8 Hz), 7.46 (s, 1H, quinoxaline-H5), 7.78 (d, 2H, Ar–H2,6, J = 8 Hz), 8.15 (d, 2H, Ar–H3,5, J = 8 Hz), 8.31 (d, 1H, quinoxaline-H8, J = 8 Hz), 11.46, 11.57 (2 s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.11 (CH2), 31.28 (CH2), 122.75, 122.83, 126.37, 127.32, 127.67, 128.11, 131.28, 131.75, 133.14, 134.95, 142.36, 143.04, 144.11 (13 Ar–C), 163.04, 164.11 (2C
O). MS m/z: 372.11 (37.12%, M+), 374.03 (12.53%, M+2), 89.07 (100). Anal. calcd for: C17H13ClN4O4 (372.76): C, 54.78; H, 3.52; N, 15.03%, found: C, 54.33; H, 3.83; N, 15.43%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.74 (t, 2H, CH2x, J = 6.4 Hz), 2.32 (t, 2H, CH2y, J = 6.4 Hz), 2.40 (s, 3H, CH3), 7.02 (d, 2H, Ar–H3,5, J = 8.4 Hz), 7.22 (s, 1H, quinoxaline-H5), 7.49 (d, 1H, quinoxaline-H7, J = 8.4 Hz), 7.68 (d, 2H, Ar–H2,6, J = 8.4 Hz), 8.03 (d, 1H, quinoxaline-H8, J = 8.4 Hz), 11.74, 11.86 (2 s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 24.55 (CH3), 29.75 (CH2), 31.19 (CH2), 123.52, 126.25, 126.44, 127.59, 130.55, 130.63, 130.67, 131.83, 132.23, 133.26, 134.26, 151.10, 151.15 (13 Ar–C), 164.44, 172.17 (2C
O). MS m/z: 341.25 (8.04%, M+), 343.09 (2.66%, M+2), 87.24 (100). Anal. calcd for: C18H16ClN3O2 (341.79): C, 63.25; H, 4.72; N, 12.29%, found: C, 63.66; H, 4.31; N, 12.53%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.72 (t, 2H, CH2x, J = 8 Hz), 2.32 (t, 2H, CH2y, J = 8 Hz), 2.41 (s, 3H, CH3), 6.65 (d, 1H, Ar–H4, J = 8 Hz), 7.21 (t, 1H, Ar–H5, J = 7.6 Hz), 7.49 (d, 1H, quinoxaline-H7, J = 8.4 Hz), 7.71 (s, 1H, Ar–H2), 7.82 (d, 1H, Ar–H6, J = 8 Hz), 7.96 (s, 1H, quinoxaline-H5), 8.09 (d, 1H, quinoxaline-H8, J = 8.4 Hz), 11.20, 11.21 (2 s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 24.17 (CH3), 29.30 (CH2), 31.26 (CH2), 123.52, 126.23, 126.43, 127.59, 130.43, 130.54, 130.97, 131.51, 132.10, 132.15, 134.31, 151.29, 151.31 (13 Ar–C), 164.15, 172.24 (2C
O). MS m/z: 341.12 (9.03%, M+), 343.40 (2.98%, M+2), 87.15 (100). Anal. calcd for: C18H16ClN3O2 (341.79): C, 63.25; H, 4.72; N, 12.29%, found: C, 63.71; H, 4.45; N, 12.66%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.73 (t, 2H, CH2x, J = 8 Hz), 2.32 (t, 2H, CH2y, J = 8 Hz), 2.40 (s, 3H, CH3), 6.77 (t, 1H, Ar–H4, J = 8 Hz), 7.83 (d, 1H, Ar–H3, J = 8.4 Hz), 7.07 (t, 1H, Ar–H5, J = 8.4 Hz), 7.13 (d, 1H, quinoxaline-H7, J = 8.4 Hz), 7.50 (d, 1H, Ar–H 6, J = 8.4 Hz), 7.53 (s, 1H, quinoxaline-H5), 8.26 (d, 1H, quinoxaline-H8, J = 8.4 Hz), 11.67, 11.77 (2s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 24.90 (CH3), 29.37 (CH2), 31.46 (CH2), 123.24, 124.35, 128.03, 129.40, 129.47, 129.60, 134.20, 134.40, 135.47, 135.60, 135.78, 137.40, 137.49 (13 Ar–C), 164.60, 168.78 (2C
O). MS m/z: 341.28 (17.51%, M+), 343.20 (5.88%, M+2), 87.04 (100). Anal. calcd for: C18H16ClN3O2 (341.79): C, 63.25; H, 4.72; N, 12.29%, found: C, 63.55; H, 4.31; N, 12.44%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.77 (t, 2H, CH2x, J = 8 Hz), 2.38 (t, 2H, CH2y, J = 8 Hz), 3.90 (s, 3H, OCH3), 7.22 (d, 2H, Ar–H3,5, J = 8.4 Hz), 7.70 (d, 1H, quinoxaline-H7, J = 8.4 Hz), 7.82 (s, 1H, quinoxaline-H5), 7.97 (d, 2H, Ar–H2,6, J = 8.4 Hz), 8.46 (d, 1H, quinoxaline-H8, J = 8.4 Hz), 11.77, 11.87 (2s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.10 (CH2), 31.15 (CH2), 55.39 (OCH3), 118.22, 119.67, 120.10, 120.46, 128.22, 128.82, 130.35, 131.15, 131.68, 132.01, 134.26, 145.03, 145.05 (13 Ar–C), 168.93, 170.68 (2C
O). MS m/z: 357.01 (12.03%, M+), 359.40 (4.11%, M+2), 82.17 (100). Anal. calcd for: C18H16ClN3O3 (357.79): C, 60.42; H, 4.51; N, 11.74%, found: C, 60.71; H, 4.82; N, 11.52%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.65 (t, 2H, CH2x, J = 8 Hz), 2.35 (t, 2H, CH2y, J = 8 Hz), 2.67 (s, 3H, COCH3), 7.21 (d, 1H, quinoxaline-H7, J = 8 Hz), 7.42 (d, 1H, quinoxaline-H8, J = 8 Hz), 7.47 (s, 1H, quinoxaline-H5), 7.76 (d, 2H, Ar–H 2,6, J = 8 Hz), 8.00 (d, 2H, Ar–H3,5, J = 8 Hz), 12.23, 12.37 (2 s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 28.22 (CO CH3), 29.82 (CH2), 31.15 (CH2), 118.03, 119.05, 120.19, 120.20, 128.39, 128.67, 130.10, 131.01, 131.26, 132.18, 134.22, 145.46, 145.55, (13 Ar–C), 167.35, 171.15 and 188.93 (3C
O). MS m/z: 369.13 (39.02%, M+), 371.17 (12.98%, M+2), 82.29 (100). Anal. calcd for: C19H16ClN3O3 (369): C, 61.71; H, 4.36; N, 11.36%, found: C, 61.35; H, 4.62; N, 11.68%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 1.62 (t, 2H, CH2x, J = 7.6 Hz), 2.24 (t, 2H, CH2y, J = 7.6 Hz), 7.27 (d, 1H, quinoxaline-H7, J = 8 Hz), 7.43 (d, 1H, quinoxaline-H8, J = 8 Hz), 7.47 (s, 1H, quinoxaline-H5), 7.97 (d, 2H, Ar–H2,6, J = 8 Hz), 8.46 (d, 2H, Ar–H 3,5, J = 8 Hz), 10.83 (s, 1H, COOH), 12.75, 12.82 (br, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.95 (CH2), 31.61 (CH2), 123.21, 125.24, 126.92, 127.30, 127.31, 127.50, 128.23, 129.66, 130.05, 130.80, 131.26, 131.27, 132.56 (13 Ar–C), 166.21, 169.31, 171.93 (3C
O). MS m/z: 371.37 (45.11%, M+), 373.17 (15.07%, M+2), 84.19 (100). Anal. calcd for: C18 H14ClN3O4 (371.77): C, 58.15; H, 3.80; N, 11.30%, found: C, 58.46; H, 3.43; N, 11.63%.
O); 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.34 (t, 3H, OCH2CH3, J = 7.2 Hz),1.73 (t, 2H, CH2x, J = 7.2 Hz), 2.34 (t, 2H, CH2y, J = 7.2 Hz), 4.36 (q, 2H, OCH2CH3, J = 7.2 Hz), 6.47 (d, 1H, quinoxaline-H7, J = 8.4 Hz), 7.41 (d, 1H, quinoxaline-H8, J = 8.4 Hz), 7.49 (d, 2H, Ar–H2,6, J = 8.4 Hz), 7.94 (s, 1H, quinoxaline-H5), 8.09 (d, 2H, Ar–H 3,5, J = 8.4 Hz), 11.89, 11.92 (2s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 14.11 (CH3), 29.40 (CH2), 31.49 (CH2), 60.03 (OCH2CH3), 117.91, 118.03, 128.24, 129.40, 129.47, 129.60, 134.11, 134.12, 135.13, 135.38, 135.39, 137.78, 137.90 (13 Ar–C), 164.07, 168.16 (2C
O of amide), 172.39 (C
O of ester). MS m/z: 399.06 (62.14%, M+), 401.19 (20.94%, M+2), 88.13 (100). Anal. calcd for: C20H18ClN3O4 (399.83): C, 60.08; H, 4.54; N, 10.51%, found: C, 60.41; H, 4.71; N, 10.77%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 2.74 (t, 2H, CH2, J = 4 Hz), 3.09 (t, 2H, CH2, J = 4 Hz), 6.86 (s, 1H, quinoxaline-H5), 6.98 (d, 1H, quinoxaline-H7, J = 4 Hz), 7.28 (d, 1H, Ar–H5, J = 8 Hz), 7.43 (s, 1H, Ar–H3), 7.62 (d, 1H, quinoxaline-H8, J = 8 Hz), 7.80 (d, 1H, Ar–H6, J = 8 Hz), 12.42, 12.43 and 12.60 (s, 3NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.75 (CH2), 31.02 (CH2), 119.12, 120.19, 120.67, 120.92, 125.45, 125.75, 126.12, 126.19, 126.67, 128.34, 129.50, 130.57, 131.02, 162.70, 167.67, 170.34 (3C
O). MS m/z (%): 438 (20.41, M+), 440.86 (6.5, M+2), 185.91 (100). Anal. calcd for: C18H13N4O3Cl3 (438): C, 49.17; H, 2.98; N, 12.74; Cl, 24.19%, found: C, 49.52; H, 2.48; N, 12.43; Cl, 24.55%.
O); 1H NMR (400 MHz, DMSO- d6) δ (ppm): 2.38 (t, 2H, CH2CH2COOH), 2.59 (t, 2H, CH2CH2COOH, J = 4 Hz), 2.64 (t, 2H, CH2, J = 4 Hz), 3.04 (t, 2H, CH2, J = 4 Hz), 7.30 (d, 1H, quinoxaline-H7, J = 8 Hz), 7.33 (s, 1H, quinoxaline-H5), 7.75 (d, 1H, quinoxaline-H8, J = 8 Hz), 10.85 (s, OH, D2O exchangeable), 12.38, 12.40 and 12.41 (s, 3NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.40 (CH2), 31.14 (CH2), 32.47 (CH2), 32.68 (CH2), 120.74, 121.01, 121.27, 124.13, 124.46, 131.20, 131.96, 161.13, 166.65, 167.04, 167.31 (4C
O). MS m/z (%): 366 (14.41, M+), 368 (4.56, M+2), 185.02 (100). Anal. calcd for: C15H15N4O5Cl (366): C, 49.12; H, 4.12; N, 15.28; Cl, 9.67%, found: C, 49.53; H, 4.37; N, 15.71; Cl, 9.32%.
O); 1H NMR (400 MHz, DMSO-d6) δ (ppm): 2.85 (t, 2H, CH2, J = 4 Hz), 3.09 (t, 2H, CH2, J = 4 Hz), 6.84 (s, 1H, quinoxaline-H5), 6.94 (d, 1H, quinoxaline-H7, J = 8 Hz), 7.28 (d, 1H, quinoxaline-H8, J = 8 Hz), 7.44 (t, 1H, Ar–H4, J = 12 Hz), 7.57 (t, 1H, Ar–H5, J = 12 Hz), 8.20 (d, 1H, Ar–H6, J = 8 Hz), 8.74 (d, 1H, Ar–H3, J = 8 Hz), 10.80 (s, OH, D2O exchangeable), 12.43, 12.44 and 12.83 (s, 3NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.66 (CH2), 31.95 (CH2), 110.45, 112.32, 117.36, 119.70, 120.73, 121.26, 124.45, 131.95, 139.66, 140.57, 142.43, 142.87, 151.11, 161.11, 168.88, 170.26 and 173.45 (4C
O). MS m/z (%): 414 (20.41, M+), 416.06 (6.5, M+2), 184.91 (100). Anal. calcd for: C19H15N4O5Cl (414): C, 55.02; H, 3.64; N, 13.51; Cl, 8.55%, found: C, 55.41; H, 3.22; N, 13.82; Cl, 8.16%.
O); 1H NMR (400 Mz, DMSO-d6) δ (ppm): 2.82 (t, 2H, CH2, J = 4), 3.03 (t, 2H, CH2, J = 4 Hz), 6.89 (s, 1H, quinoxaline-H5), 6.97 (d, 1H, quinoxaline-H7, J = 8 Hz), 7.32 (d, 1H, quinoxaline-H8, J = 8 Hz), 7.48 (t, 1H, Ar–H4, J = 8 Hz), 7.60 (t, 1H, Ar–H5, J = 8 Hz), 8.28 (d, 1H, Ar–H6, J = 8 Hz), 8.78 (d, 1H, Ar–H3, J = 8 Hz), 12.22 and 12.54 (s, 2NH, D2O exchangeable). 13C NMR (DMSO-d6) δ (ppm): 29.33 (CH2), 31.54 (CH2), 115.66, 115.72, 115.85, 115.94, 117.59, 119.53, 120.41, 121.23, 121.31, 124.07, 131.38, 135.81, 135.84 (13 Ar–C), 159.68, 165.73, 166.70 and 169.74 (4C
O). MS m/z (%): 396 (9.05, M+), 398.52 (2.98, M+2), 102.34 (100). Anal. calcd for: C19H13N4O4Cl (396): C, 57.51; H, 3.30; N, 14.12; Cl, 8.94%, found: C, 57.04; H, 3.72; N, 14.43; Cl, 8.56%.
VEGFR-2 inhibition estimation: VEGFR-2 inhibition estimation was executed utilizing VEGFR-2 (KDR) Kinase Assay Kit of Biosciences and the IC50 values were detected according to the manufacturer's instructions and the reported method.37
Quantitative determination of VEGFR2 in MCF-7 cell line: ELISA analysis to determine VEGFR2 quantitatively in MCF-7 cell line is following the reported procedure38
Effect of 14 on angiogenesis (VEFG-A) level: ELISA analysis to determine VEGF-A secretion is following the reported procedure.40
Cell cycle analysis: FACS Caliber flow cytometer was utilized to determine the effect of 14 on the cell cycle of breast tumor NCF-7 guided by the previously reported method.39
Annexin-V-FITC apoptosis assay: the apoptosis of 14 was detected through Annexin V-FITC/PI apoptosis detection kit utilizing FACS Caliber flow cytometer following the reported procedure.40
Effect of 14 on active caspase-3 level
Caspase-3 is evaluated by using the reported method.41
Evaluation of apoptosis regulator p53
P53 expression is measured in MCF-7 by the reported method.42
Evaluation of BAX and BCL2 levels43,44
Footnote |
| † Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d3ra05066a |
| This journal is © The Royal Society of Chemistry 2023 |