Next Article in Journal
Deposition of Hybrid Photocatalytic Layers for Air Purification Using Commercial TiO2 Powders
Previous Article in Journal
Activation and Identification of a Griseusin Cluster in Streptomyces sp. CA-256286 by Employing Transcriptional Regulators and Multi-Omics Methods
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pyrrolizine/Indolizine-NSAID Hybrids: Design, Synthesis, Biological Evaluation, and Molecular Docking Studies

by
Mohammed A. S. Abourehab
1,*,
Alaa M. Alqahtani
2,
Faisal A. Almalki
2,
Dana M. Zaher
3,
Ashraf N. Abdalla
4,5,
Ahmed M. Gouda
6,* and
Eman A. M. Beshr
7
1
Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
2
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
3
Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
4
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
5
Department of Pharmacology, Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum 2404, Sudan
6
Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
7
Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(21), 6582; https://doi.org/10.3390/molecules26216582
Submission received: 10 October 2021 / Revised: 27 October 2021 / Accepted: 27 October 2021 / Published: 30 October 2021

Abstract

:
In the current study, eight new hybrids of the NSAIDs, ibuprofen and ketoprofen with five pyrrolizine/indolizine derivatives were designed and synthesized. The chemical structures of these hybrids were confirmed by spectral and elemental analyses. The antiproliferative activities of these hybrids (5 μM) was investigated against MCF-7, A549, and HT-29 cancer cell lines using the cell viability assay, MTT assay. The results revealed 4–71% inhibition of the growth of the three cancer cell lines, where 8a,e,f were the most active. In addition, an investigation of the antiproliferative activity of 8a,e,f against MCF-7 cells revealed IC50 values of 7.61, 1.07, and 3.16 μM, respectively. Cell cycle analysis of MCF-7 cells treated with the three hybrids at 5 μM revealed a pro-apoptotic increase in cells at preG1 and cell cycle arrest at the G1 and S phases. In addition, the three hybrids induced early apoptotic events in MCF-7 cells. The results of the molecular docking of the three hybrids into COX-1/2 revealed higher binding free energies than their parent compounds 5a,c and the co-crystallized ligands, ibuprofen and SC-558. The results also indicated higher binding free energies toward COX-2 over COX-1. Moreover, analysis of the binding modes of 8a,e,f into COX-2 revealed partial superposition with the co-crystallized ligand, SC-558 with the formation of essential hydrogen bonds, electrostatic, or hydrophobic interactions with the key amino acid His90 and Arg513. The new hybrids also showed drug-likeness scores in the range of 1.06–2.03 compared to ibuprofen (0.65) and ketoprofen (0.57). These results above indicated that compounds 8a,e,f deserve additional investigation as potential anticancer candidates.

1. Introduction

Currently, more than 150 anticancer drugs approved by the FDA for the treatment of different types of cancer are available in the market [1]. However, the rates of morbidity and mortality due to cancers are still high, even within high-income countries [2]. In addition, the low rate of survival in patients with metastatic cancers [3] also confirms the urgent need for continuous research in this field.
Treatment of cancer with a single drug was associated in many cases with the development of resistance and low therapeutic outcomes [3,4]. To overcome this problem, several approaches have been developed in cancer therapy. Among these approaches, combination therapy was emerged as a promising approach in cancer therapy [5]. The combined drugs can be used to target different essential pathways in cellular proliferation, where they can act either synergistically or additively to combat the overgrowth of cancer [6,7].
Combination therapy has showed preferential advantages over the single-agent treatment for survival rates and the tumor response [8]. Due to the promising outcomes of combination therapies, a huge number of clinical trials were launched to investigate combination therapies in cancer and other diseases [9]. However, combination therapy also has several disadvantages. Among these, the higher side effects which develop after administration of two or more drugs compared to the single-agent treatment [10]. Combination therapy also produced higher toxicity than the single agent treatment [8]. Pharmacokinetic problems may also appear on administration of combined drugs since management of the pharmacokinetics of a single drug is easier. In addition, the potential for drug-drug interactions in combination therapy is higher than the single-agent treatment [11].
Hybrid/conjugate drugs also emerged as a promising strategy in cancer therapy [12,13]. They could provide a solution to the poor patient acceptability to combination therapy [13,14]. The hybrid/conjugate drugs could also provide several advantages over the traditional combination therapy including the lower toxicity and side effects [15,16]. In addition, the management of the pharmacokinetics of a single agent could be easier.
NSAIDs are also considered one of the cancer pain medications [17]. They constitute a key component of the WHO analgesic leader to control cancer pain [18]. However, besides their use as analgesic anti-inflammatory agents, NSAIDs have also displayed a chemopreventive effect in colorectal and breast cancer [19,20].
Our literature review revealed several acetylsalicylic acid-metal complexes with potent anticancer activity. The Se-aspirin complex I exhibited potent anticancer activity against a panel of 6 cancer cell lines with IC50 values in the range of 1.3–4.4 μM [21], Figure 1. Mechanistic studies of I into colorectal cancer cells revealed cell cycle arrest at G1 and G2/M phases and induction of apoptosis. In addition, Weninger et al. [22] have reported a series of acetylsalicylic acid derivatives with Zeise’s Salt (IIa–d). These derivatives exhibited growth-inhibitory activities against HT-29 and MCF-7 cancer cells at IC50 values in the range of 30-50 μM. In addition, Baecker et al. [23] have reported a series of cobalt alkyne complexes with fluorinated acetylsalicylic acid III. These complexes showed cytotoxic and apoptotic activities against the COX-1/2-expressing cancer cell lines (HT-29 and MDA-MB-231). On the other hand, compounds III exhibited much lower activity against MCF-7 cells which lack COX protein.
In addition, several successful examples of NSAIDs hybrids/conjugates in cancer research have been also reported, Figure 1. Although the parent NSAIDs have showed weak cytotoxic activities [24,25,26,27], their hybrids with the cytotoxic agents were reported to have potent anticancer activity. Among these hybrids, the riboflavin-dexibuprofen conjugates IVa–e were synthesized by Banekovich et al. [24]. A biological evaluation of these conjugates IVa–e revealed growth-inhibitory activity of MCF-7 and HT-29 at IC50 values of 8–15 µM. These results indicated that the conjugates have much higher antiproliferative activities than their parent components, dexibuprofen and tetraacetylriboflavin.
Curci et al. [25] have synthesized prodrug conjugate V of kiteplatin Pt(IV) and two molecules of ibuprofen. The new conjugate exhibited much higher cytotoxic activity compared to the parent components. It inhibited the growth of HCT15 and HCT116 cancer cell lines at IC50 of 0.45 and 0.26 µM, respectively.
The ibuprofen-podophyllotoxin conjugate VI was reported among a small series of podophyllotoxin designed by Zhang et al. [26]. Evaluation of cytotoxicity of VI against Bel-7402 and Bel-7402/5-FU cancer cell lines revealed IC50 values of 18.88 and 10.28 µM, respectively. These results indicated higher cytotoxic potential toward the resistant cancer cell line.
The hybrid VII was designed by combining 5,16-pregnadiene and ibuprofen [27]. Evaluation of the antiproliferative activity of VII revealed higher growth-inhibitory activity against the human anaplastic astrocytoma (U373) cells than the parent compounds.

Rational Design

Previously, we reported compound VIIIa–c (Figure 2) with in vivo anti-inflammatory activities comparable with that of ibuprofen [28,29]. Only two of these hybrids (VIIIb,c) were also investigated for their cytotoxic activity against three cancer cell lines (IC50 = 0.06–0.87 μM). These results motivated us to investigate the structure-activity relationship and the potential mechanism which mediates the cytotoxic activity of this scaffold.
Many of the NSAIDs displayed weak antiproliferative activity which could be COX-dependent or independent [30]. Esterification/amidation of the carboxylic acid group in ibuprofen was associated in some cases with increases in the anti-inflammatory activity and decrease in GIT side effects [31,32]. Some of the ester and amide derivatives of ibuprofen also showed improved antiproliferative activity compared to the parent drug [30]. On a cellular level, this improvement of the antiproliferative activity could be due to the increased lipophilicity and enhancer cellular uptake [33].
Encouraged by the above findings, we have designed a new series of hybrids (scaffold B) by incorporating the pyrrolizine/indolizine derivatives (scaffold A) with NSAIDs, Figure 3. Ibuprofen was selected as the NSAID component of the new hybrids based on the promising cytotoxic activity of VIIIb,c. In addition, ketoprofen was also selected in this study to compare the effect of the two drugs on the antiproliferative activity of the new hybrids. The aim of this selection was to compare the impact of the two drugs on cytotoxic activities of the final hybrids, Figure 3.
To investigate the relationship between the chemical structure and the antiproliferative activity of the new hybrids (scaffold B), several pyrrolizine/indolizine derivatives (scaffold A) were used. In addition, the new hybrids were designed bearing electron-donating (OCH3) and electron-withdrawing (F, Cl, and Br) substituents at the para-position of ring A to compare the impact of the electronic effects of their antiproliferative activity, Figure 3. The new hybrids will be evaluated for their antiproliferative activity against a panel of cancer cell lines. An additional investigation will also be performed to evaluate the potential mechanism of their action of the most active compounds.

2. Results and Discussion

2.1. Chemistry

The synthesis of the target hybrids 8a–i is presented in Scheme 1. Compounds 2a,b were obtained from the reaction of 1a,b with malononitrile according to the previous reports [34,35]. In addition, compounds 4a–d were prepared from the reaction of chloroacetyl chloride with the appropriate aniline derivatives 3a–d [34,35]. On the other hand, the pyrrolizine/indolizine derivatives 5a–e were prepared from the reaction of 2a,b with the appropriate acetanilide derivatives 4a–d [36].
To prepare 4-methoxy/fluoro indolizine derivatives, 2b was also refluxed with the appropriate acetanilide 4a,b for 48 h. However, the duplication of the reflux time was also associated with the formation of uncyclized derivatives rather than the target indolizines. The IR spectra of the obtained products revealed absorption bands of the geminal cyano groups, which confirms the formation of uncyclized products. These results align with the results of our previous report [35].
Preparation of the target hybrids 8a–i was achieved in two steps following the previous report [29]. In the first step, the NSAIDs, ibuprofen and ketoprofen were converted into their acid chloride derivatives 7a,b using thionyl chloride. The second step takes place through an acylation of the primary amino groups in compounds 5a–e using the freshly prepared acid chloride derivatives 7a,b. Structural elucidation of the new hybrids 8a–i was performed. The spectral data of compounds 8a,b is discussed below.
The IR spectrum of pyrrolizine-ibuprofen 8a revealed absorption bands at 2221 and 1659 cm−1 indicate the cyano and carbonyl groups, respectively (Supplementary data, Figure S1). The 1H-NMR spectrum of 8a revealed seven signals at the aliphatic region (δ 0.88–4.29 ppm) which indicate the aliphatic protons, (Supplementary data, Figures S10–S13). The two methyl groups of the isobutyl moiety (-CH2CH(CH3)2) appears as a doublet signal at δ 0.88 ppm. Another doublet signal at δ 1.61 indicates the methyl group of the propionamide moiety (-COCHCH3). In addition, four doublet signals are found at δ 6.74, 7.04, 7.21, and 7.26 ppm indicate the two para-substituted phenyl rings. Two singlet signals at δ 8.21 and 9.03 ppm indicate the two amide protons.
On the other hand, the IR spectrum of the ketoprofen hybrid 8b revealed absorption bands at 2211 and 1698 cm−1, indicating the cyano and carbonyl groups, respectively (Supplementary data, Figure S2). A total of 6 signals at the aliphatic region (δ 1.62–4.27 ppm) in the 1H-NMR spectrum of 8b are observed indicating the aliphatic protons, (Supplementary data, Figures S20–S23). The signal corresponding to the methyl group of the propionamide moiety (-NHCOCHCH3) appears as a doublet signal at δ 1.62 ppm. The spectrum also shows one triplet signal at δ 2.83, which indicates the methylene group (CH2-1) of the pyrrolizine nucleus. A multiplet at δ 6.69–7.68 ppm indicates 12 of the aromatic protons of the 3 phenyl rings, while a single signal at δ 7.86 ppm indicates the aromatic proton at ortho-position of the phenyl propionamide ring (B). In addition, 2 singlet signals at δ 8.94 and 9.02 ppm indicates the two amide protons in this compound.
Supplementary data including all spectral data and copies of IR, 1H-NMR, and 13C-NMR spectra of the final compounds 8a–i are provided with this manuscript (Supplementary data, Figures S1–S78).

2.2. Biological Evaluation

2.2.1. Antiproliferative Activity

Screening Assay

Previously, the ibuprofen hybrids/conjugates IVVIII exhibited antiproliferative activities against different types of cancer cell lines [24,25,26,27,28,29]. Accordingly, the new hybrids 8a–i were evaluated for their antiproliferative activity using the cell viability assay, MTT assay following the previous report [37]. In this assay, the new hybrids were evaluated against three cancer cell lines including MCF-7 breast, A549 lung, and HT-29 colon cancer cell lines. The antiproliferative activities of the starting materials 5a–e, ibuprofen and ketoprofen, were also investigated in this assay. The cancer cells were treated with the test compounds at a single dose (5 μM) for 72 h. The inhibition in the viability of the cancer cell lines was calculated relative to the untreated control. The results (growth%) are represented in Figure 4.
The results of the cell viability assay of the tested hybrids against MCF-7 cells revealed 12–71% inhibition of the cancer cells’ viability, Figure 4, which were much higher compared with those of ibuprofen (5%) and ketoprofen (9%). The changes in the cell viability of MCF-7 cells treated by all the new hybrids 8a–i were statistically different from the control (p < 0.01). Compounds 8a,e,f induced more than 50% inhibition of the growth of MCF-7 cells at 5 μM. In addition, the pyrrolizine derivatives 8a–f showed higher growth-inhibitory activities than their parent compounds 5a–c, however, the indolizine derivatives 8g–i exhibited lower growth-inhibitory activities than their parent compounds 5c,d. The results also indicated higher antiproliferative for the ibuprofen derivatives (8a,c,g) compared to their corresponding ketoprofen analogs (8b,d,h). Among the new hybrids, 8f exhibited the highest antiproliferative activity (71%) against MCF-7 cells, which was statistically different from those of the control and ibuprofen (p < 0.01).
The effects of the new hybrids 8a–i on the growth of A549 cancer cell line are presented in Figure 5. Except for 8h, the new hybrids exhibited 13–48% inhibition in the growth of A549 cells compared to the control (p < 0.01). The results also revealed higher antiproliferative activity for the ibuprofen hybrids than the ketoprofen analogs. However, no antiproliferative activity was observed for ibuprofen and ketoprofen against A549 cells. Among the new hybrids, 8a,e,f showed the highest antiproliferative activity.
Finally, compounds 8a–i were also investigated for their effects on the viability of HT-29 cancer cells. The results are presented in Figure 6. Except for compound 8b, all of the tested hybrids included 4–53% inhibition in the viability of HT-29 cells. However, both ibuprofen and ketoprofen did not show antiproliferative activity against HT-29 cells at 5 μM.
The results also indicated higher antiproliferative activity for the ibuprofen derivatives (8a,c,e,g) against HT-29 cells than their ketoprofen analogs (8b,d,f,h). Among the new hybrids, 8a,e,f exhibited the highest antiproliferative activity (47–49%) against HT-29 cells compared to the control (p < 0.01).
In conclusion, the results of the cell viability assay showed the highest sensitivity for MCF-7 cells toward the new hybrids. Moreover, compounds 8a,e,f were the most active in inhibiting the proliferation of MCF-7 cells.

Cytotoxicity Assay

Based on the results of the MTT viability assay (Figure 4, Figure 5 and Figure 6), three of the new hybrids (8a,e,f) were selected for further investigation of their antiproliferative activities. The MTT assay was used to determine the IC50 values of the selected hybrids following the previous report [38]. MCF-7 cell line, the most sensitive to the antiproliferative effect of the new hybrids, was selected for this study. Doxorubicin was used as a reference drug. The cancer cells were treated with the test compounds at different concentrations. The results expressed as IC50 values were determined, Table 1.
The results of the MTT assay revealed that the three hybrids inhibit the growth of MCF-7 cells at IC50 values in the range of 1.07–7.61 μM compared to doxorubicin (IC50 = 2.07 μM). The IC50 value of doxorubicin was slightly higher than the reported value [39]. Among the three compounds, 8e was the most active in inhibiting the growth of MCF-7 cells, while 8a showed the lowest cytotoxicity.

2.2.2. Cell Cycle Analysis

Previous reports indicated that the amide derivatives of ibuprofen/ketoprofen induced cell cycle arrest at the G0/G1 or G1/S phases in cancer cell lines [30].
In the current study, the effect of compounds 8a,e,f on the cell cycle distributions of MCF-7 cells was investigated 8a,e,f were also investigated. The cancer cells were treated with the test compounds at 5 μM for 48 h. This study was performed following the previous report [36]. The results are presented in Figure 7.
The result of cell cycle analysis revealed that each of the 3 compounds showed profound increase in the preG1 phase in MCF-7 cells, as an indication of their pro-apoptotic activities, especially 8e that showed 10-fold increase compared to control, Table 2. Additionally, 8a and 8e arrested cells in the S phase at the expense of G1 and G2/M phases, while 8f slightly increased cells in the G1 phase.

2.2.3. Annexin V-FITC/PI Apoptosis Assay

The amide derivatives of ibuprofen/ketoprofen were also reported to induce apoptosis in different types of cancer cell lines [30]. In the current study, annexin V-FITC/PI double staining protocol was used to investigate the effects of compounds 8a,e,f on apoptotic events in MCF-7 cells. The cancer cells were treated with each of the 3 hybrids at 5 μM for 48 h. The study was performed following the previous report [36]. The results were presented in Figure 8.
The 3 hybrids 8a,e,f induced increase in the early apoptotic events in MCF-7 cells ranging from 18–50 folds (11–30%) compared to the control, Table 3. This was at the expense of late and necrotic events which were absent.

2.3. Computational Studies

2.3.1. Molecular Docking Studies

The main mechanism of action that mediate the anti-inflammatory activity of ibuprofen/ketoprofen is the reversible inhibition of COX-1/2 isoenzymes [40,41]. In addition, the mechanistic studies of the starting material 5c revealed moderate inhibition of COX-1/2 with weak selectivity toward COX-2 [28]. Although several reports indicated that the antiproliferative activity of NSAIDs are COX-independent, some other reports described a correlation between COXs inhibition and this activity [30]. Moreover, many of the NSAIDs prodrugs have also displayed intrinsic anti-inflammatory activities mediated even in part by COXs inhibition [42].
Based on the above mentioned finding, we performed a molecular docking study of compounds hybrids 8a,e,f into the two COXs to compare the binding characteristics against those of their parent compounds (5a,c and ibuprofen). The docking study was performed into the crystal structure of COX-1 (pdb 1EQG) [43] and COX-2 (pdb 1CX2) [44] using AutoDock 4.2 [45]. The ligands (8a,e,f) and protein molecules were prepared into the suitable formate (pdbqt) for AutoDock following the previous report [46]. In the current study, COX-1/2 proteins were used as rigid molecules, while the ligands were docked as flexible molecules. The 2/3D binding modes of the tested compounds were generated by Discovery Studio Visualizer [47].
Firstly, ibuprofen was re-docked into the active site of COX-1 to validate the docking procedures. The 2/3D binding modes of the re-docked ibuprofen was investigated against those of the co-crystallized ligand. A RMSD of 0.87 Å was observed between the re-docked and the co-crystallized ligand. The re-docked molecule showed identical hydrogen bonds and hydrophobic interactions with those of the co-crystallized ibuprofen, Supplementary data, Figure S79.
Validation of the docking study into COX-2 (pdb: 1CX2) was also performed. the co-crystallized ligand, SC-558 was docked into the active site of COX-2. The results revealed superposition of the re-docked SC-558 with the co-crystallized ligand with RMSD of 1.30 Å. The binding mode of the re-docked SC-558 overlaid with the co-crystallized ligand is shown in Supplementary data, Figure S80.
The results of the docking study of the 3 hybrids (8a,e,f) into COX-1 binding free energies in the range of −10.36 to −10.55 kcal/mol compared to ibuprofen (energy (ΔGb = −8.43 kcal/mol), Table 4. On the other hand, they exhibited higher binding free energies toward COX-2 (ΔGb = −10.70 to −12.56 kcal/mol) compared to −10.78 kcal/mol for SC-558. The binding free energies of the three hybrids were also higher than their parent compounds 5a,c.
Compound 8a exhibited a binding free energy (ΔGb ) of -10.36 kcal toward COX-1, which was higher than ibuprofen, Table 4. An investigation of the binding mode of 8a revealed that the best fitting conformation adopted a binding orientation superposing with the co-crystallized ibuprofen into COX-1, where the 4-methoxyaniline moiety in 8a superposed with the 4-isobutylphenyl moiety in ibuprofen, Figure 9. In addition, the carboxamide moiety at C2 of compound 8a was also superposed with the propionic acid moiety in ibuprofen. This orientation allowed 8a to interact similarly to ibuprofen with Arg120 and Tyr355 in COX-1 forming a cluster of four conventional hydrogen bonds. Compound 8a and ibuprofen exhibited similar hydrophobic interactions with Val116, Val349, Ala527, and Leu531.
The results of the docking study of compound 8a into COX-2 also revealed a higher binding free energy than SC-558. These results indicated that the hybrid 8a has higher affinity toward COX-2 over COX-1. An analysis of the binding mode of the best fitting conformation of compound 8a into COX-2 revealed nice superposition of the two phenyl rings with the phenyl rings of SC-558, Figure 10. The pyrrole ring in 8a was also located near the binding position of the pyrazole ring. Compound 8a exhibited one conventional hydrogen bond with Arg120 and one carbon hydrogen bond with Ser353. In addition, 8a showed one electrostatic interaction with Arg513. Based on these results, the higher binding free energy of 8a toward COX-2 could be attributed to the electrostatic interaction with Arg513 and the multiple hydrophobic interactions with the hydrophobic residues in COX-2.
Compound 8e also showed higher affinity toward COX-1 than ibuprofen. However, 8e did not show any conventional hydrogen bonds with the amino acids in COX-1, Figure 11.
The results of the docking study of 8e into COX-2 also revealed higher binding free energy compared to COX-1. The 4-bromophenyl moiety in 8e superposed with the bromophenyl moiety in SC-558 into the hydrophobic pocket of COX-2, Figure 12. In addition, the pyrrolizine nucleus was also overlaid with the pyrazole ring in SC-558, while the phenyl propionamide in 8e extended with the phenyl sulfonamide moiety in SC-558 into the side pocket of COX-2. Compound 8e displayed six hydrogen bonds including four conventional hydrogen bonds with His90, Leu352, and Arg513 and two carbon hydrogen bonds with His90 and Gly526, Figure 12.
Among all the tested compounds, 8f displayed the highest binding free energy toward COX-2, Table 4. An investigation of the binding interactions of 8f revealed one conventional hydrogen bond with Arg120 and one carbon hydrogen bond with Val116, Figure 13.
The orientation of the best fitting conformation of 8f into COX-2 also revealed partial overlay of the two phenyl rings with the two phenyl rings in SC-558, Figure 14. The bromophenyl moiety in 8f occupied the hydrophobic pocket of COX-2 and formed identical hydrophobic interactions with Leu384, Tyr385, and Trp387. On the other hand, the superposition of the phenyl carboxamide at C2 in 8f with the phenyl sulfonamide moiety in SC-558 into the side pocket of COX-2 allowed the two molecules to form one electrostatic interaction with His90 and two hydrophobic interactions with Ser353 and Val523. Moreover, 8f showed one carbon hydrogen bond with Arg120 compared to unfavorable donor-donor interaction for SC-558.
The 2/3D binding modes of the parent compounds (5a,c) were illustrated in Supplementary data, Figures S81 and S82.
In conclusion, the three hybrids 8a,e,f exhibited higher free energies toward COX-1/2 than the parent compounds (5a,c) and the co-crystallized ligands (ibuprofen and SC-558). In addition, their binding free energies toward COX-2 were higher than those for COX-1. An analysis of the binding modes of these hybrids into COX-2 revealed partial superposition with SC-558 and formation of hydrogen bonds, electrostatic, or hydrophobic interactions with the key amino acids His90, and Arg513.

2.3.2. Drug-Likeness and ADME Studies

Pharmacokinetic properties play a critical role in the discovery and development of the new drug candidates [48]. The successful transition of these new compounds to the development stage depends mainly on their pharmacokinetics and pharmacodynamics properties [49]. In this study, the molecular properties related to the pharmacokinetic and drug-likeness parameters of compounds 8a–i were evaluated and compared with those of ibuprofen, ketoprofen, and compounds 3. The study of the molecular properties was calculated using SwissADME [50] and Molsoft L.L.C. The results are presented in Table 5.
Based on Lipinski’s rule, an orally active drug has a total number of hydrogen bond donors ≤ 5, hydrogen bond acceptors ≤ 10, logP < 5, and molecular weight < 500 da [51]. An investigation of physicochemical parameters of 8a–i revealed that their molecular weights are above 500 daltons. However, the molecular weights are less than that of compound VI. The polar surface areas of 8a–i are either comparable or lower than that of compound VI. The calculated logP values of the nine hybrids are in the range of 2.12–3.73 compared to compound VI (logP = 3.71). The results also showed that compound 8a,c have no violation from Lipinski’s rule, while all of the remaining compounds showed only one violation (MW > 500).
The tested hybrids also showed drug-likeness scores (DLS) in the range of 1.06−2.03 compared to 1.01 for compound VI. The fraction of 8a–i that can undergo GIT absorption was calculated following the previous report [52]. The results revealed that 80.10–84.75% of 8a–i could be absorbed from GIT compared to compound VI (80.40%).
The detailed results of the ADME study are provided in Supplementary data, Tables S1–S18.

3. Conclusions

The design of anticancer-NSAID hybrids could be used to avoid some of the problems encountered in combination therapy. Many of the hybrids combining ibuprofen with anticancer agents have displayed promising in vitro cytotoxic activities. In the current study, eight hybrids bearding the NSAIDs (ibuprofen and ketoprofen) with five pyrrolizine/indolizine derivatives (5a–e) were designed and synthesized. A structural elucidation of the new hybrids was confirmed using IR, mass, 1H-NMR, 13C NMR and DEPT C135 spectra. The new hybrids were evaluated for their antiproliferative activities against three (MCF-7, A549, and HT-29) cancer cell lines using a quick screening assay. The results revealed a 4–71% inhibition in the growth of the cancer cells, where MCF-7 cells were the most sensitive. Based on the results of this assay, compounds 8a,e,f, the most active against MCF-7 cells were selected for additional biological evaluations. The cytotoxic activity of 8a,e,f against MCF-7 cells revealed IC50 values in the range of 1.07–7.61. Cell cycle analysis of MCF-7 cells treated with the three hybrids at 5 μM revealed pro-apoptotic increase in cells at preG1 and cell cycle arrest at the G1 and S phases. In addition, the three hybrids induced early apoptotic events in MCF-7 cells. A molecular docking study of the three hybrids into the active sites of COX-1/2 was also performed. The results revealed higher binding free energies for the three hybrids toward COX-1/2 compared to the parent compounds 5a,c and the co-crystallized ligands, ibuprofen and SC-558. The results also indicated the higher binding free energies of the three hybrids toward COX-2 over COX-1. An analysis of the binding modes of the best fitting conformation of the three hybrids into COX-2 revealed partial superposition with SC-558 and formation of hydrogen bonds, electrostatic, and hydrophobic interactions with the key amino acid such as His90, and Arg513. The new hybrids also showed drug-likeness score of 1.06–2.03. The above results support the future investigation of compounds 8a,e,f as potential anticancer candidates.

4. Experimental Protocol

4.1. Chemistry

Chemical reagents and solvents were obtained from commercial sources. Solvents are dried by standard methods when necessary. The purity of the new compounds was checked with TLC. Melting points (m.p.) are uncorrected and were determined by IA 9100MK-Digital melting point apparatus (Cole-Parmer, East Bunker Ct Vernon Hills, IL, USA). Infrared spectra (IR) were recorded using BRUKER TENSOR 37 spectrophotometer (Bruker, Billerica, MA, USA). The proton magnetic spectra were recorded on BRUKER AVANCE III at 500 MHz, 13C NMR (125 MHz), and DEPT C135 (125 MHz). Mass spectra were recorded on Agilent UPLC/MS/MS 1260 infinity II with 6420 Triple quad LC/MS detector at Faculty of Pharmacy, Minia University, Minia, Egypt.
Compounds 2a,b [34], and 4a–d [35], and 5a–e [36] were prepared according to the previous reports.

4.1.1. General Procedure (A) for Preparation of Compounds (8a–i)

The synthesis of the hybrids 8a–i was achieved using the same reaction conditions applied in the synthesis of compound 8e [28].

Preparation of Compounds 7a,b

Thionyl chloride (1 gm, 8.41 mmol) was added to 5.9 mmol of (±)-ibuprofen/ketoprofen in a dry round-bottom flask. The reaction mixture was refluxed on a steam bath. After for 2 h reflux, the excess thionyl chloride was removed under reduced pressure. The residue (ibuprofen/ketoprofen acetyl chloride) was dissolved in methylene chloride (30 mL).

Preparation of Compounds 8a–j

A solution of compound 5a–e (3 mmol) in methylene chloride (30 mL) was added dropwise to the acid chloride solution obtained from the appropriate NSAIDs. To the reaction mixture, 0.5 mL of TEA was added, and the mixture was stirred for 2 h at rt. The reaction mixture was left to stand overnight. The solvent was evaporated under reduced pressure and the solid residue obtained was washed with 10 mL of aqueous sodium carbonate 2% and left to dry. The dry residue was recrystallized from ethanol-acetone.
(R,S)-7-Cyano-6-(2-(4-isobutylphenyl)propanamido)-N-(4-methoxyphenyl)-2,3-dihydro-1H-pyrrolizine-5-carboxamide (8a) The title compound was prepared from the reaction of compound 5a (0.89 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ibuprofen (1.22 g, 5.9 mmol) according to the general procedure A. Compound 8a was obtained as white solid product, m.p. 232–4 °C, yield 67%. IRυmax/cm−1 3409, 3268 (NHs), 2952, 2868, 2832 (C-H aliphatic), 2221 (CN), 1659 (C=O), 1601, 1544, 1511 (C=C, C=N), 1425, 1308, 1248 (C-N, C-O). 1H-NMR (DMSO-d6, 500 MHz, δ ppm): δ 0.88 (d, 6H, J = 6.2 Hz, CH(CH3)2), 1.61 (d, 3H, J = 6.6 Hz, CHCH3), 1.77–1.85 (m, 1H, CH(CH3)2), 2.39–2.45 (m, 4H, pyrrolizine CH2-2 + Ph-CH2), 2.84 (t, 2H, J = 7.2 Hz, pyrrolizine CH2-1), 3.79 (s, 3H, OCH3), 4.16–4.29 (m, 3H, CHCH3+pyrrolizine CH2-3), 6.74 (d, 2H, J = 7.8 Hz, Ph (A) CH-3+CH-5), 7.04 (d, 2H, J = 7.02 Hz, Ph (B) CH-3+CH-5), 7.21 (d, 2H, J = 7.8 Hz, Ph (A) CH-2+CH-6), 7.26 (d, 2H, J = 7.1 Hz, Ph (B) CH-2+CH-6), 8.21 (s, 1H, CONHCH), 9.03 (s, 1H, CONHPh). 13C-NMR (DMSO, 125 MHz, δ ppm): δ 18.35, 22.39, 24.79, 25.45, 30.15, 45.03, 46.52, 49.41, 55.42, 84.44, 113.92, 114.11, 119.83, 121.29, 124.84, 127.38, 129.79, 130.82, 137.08, 141.13, 145.60, 146.13, 157.48, 176.88. DEPT C135 (DMSO, 125 MHz, δ ppm): δ 18.35, 22.39, 24.79, 25.46, 30.16, 45.03, 46.51, 49.41, 55.42, 113.91, 121.28, 127.38, 129.79. LC-MS: m/z 471.40 [M-13]+. Anal. Calcd. for C29H32N4O3 (484.59): C, 71.88; H, 6.66; N, 11.56. Found C, 71.37; H, 6.42; N, 10.70.
(R,S)-6-(2-(3-Benzoylphenyl)propanamido)-7-cyano-N-(4-methoxyphenyl)-2,3-dihydro-1H-pyrrolizine-5-carboxamide (8b) The title compound was prepared from the reaction of compound 5b (0.89 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ketoprofen (1.45 g, 5.9 mmol) according to the general procedure A. Compound 8b was obtained as white solid product, m.p. 246–8 °C, yield 64%. IRυmax/cm−1 3284, 3203 (NHs), 3085, 3003, 2968, 2841 (C-H aliphatic), 2211 (CN), 1698 (C=O), 1587, 1555, 1508 (C=C, C=N), 1429, 1302, 1230 (C-N, C-O). 1H-NMR (CDCl3, 500 MHz, δ ppm): δ 1.62 (d, 3H, J = 7.1 Hz, CHCH3), 2.38-2.43 (m, 2H, pyrrolizine CH2-2), 2.83 (t, 2H, J = 7.5 Hz, pyrrolizine CH2-1), 3.71 (s, 3H, OCH3), 3.99 (q, 1H, J = 6.7 Hz, CHCH3), 4.17–4.27 (m, 2H, pyrrolizine CH2-3), 6.69 (d, 2H, J = 8.3 Hz, aromatic Hs), 7.17 (d, 2H, J = 8.3 Hz, aromatic Hs), 7.31 (t, 1H, J = 7.5 Hz, aromatic H), 7.42 (t, 2H, J = 7.3 Hz, aromatic Hs), 7.55 (t, 2H, J = 8.1 Hz, aromatic Hs), 7.64 (d, 1H, J = 7.5 Hz, aromatic H), 7.68 (d, 2H, J = 7.5 Hz, aromatic Hs), 7.86 (s, 1H, Ph (B) CH-2), 8.94 (s, 1H, CONHCH), 9.02 (s, 1H, CONHPh). 13C-NMR (CDCl3, 125 MHz, δ ppm): δ 18.45, 24.79, 25.47, 46.49, 49.41, 55.40, 84.54, 113.96, 114.18, 119.73, 121.12, 124.90, 128.32, 128.76, 129.15, 129.55, 130.09, 130.76, 131.62, 132.64, 137.23, 137.94, 140.74, 145.72, 156.29, 157.47, 176.03, 196.78. DEPT C135 (CDCl3, 125 MHz, δ ppm): δ 18.46, 24.79, 25.47, 46.48, 49.41, 55.40, 113.96, 121.11, 128.33, 128.76, 129.15, 129.56, 130.10, 131.63, 132.65. LC-MS: m/z 531.40 [M-H]+. Anal. Calcd. for C32H28N4O4 (532.59): C, 72.16; H, 5.30; N, 10.52. Found C, 71.84; H, 4.98; N, 11.05.
(R,S)-7-Cyano-N-(4-fluorophenyl)-6-(2-(4-isobutylphenyl)propanamido)-2,3-dihydro-1H-pyrrolizine-5-carboxamide (8c) The title compound was prepared from the reaction of compound 5b (0.85 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ibuprofen (1.22 g, 5.9 mmol) according to the general procedure A. Compound 8c was obtained as white solid product, m.p. 239–41 °C, yield 56%. IRυmax/cm−1 3408, 3259 (NHs), 3065, 3046, 3013, 2949 (C-H aliphatic), 2223 (CN), 1662 (C=Os), 1612, 1548, 1510 (C=C, C=N), 1466, 1425, 1317, 1277 (C-N, C-O). 1H-NMR (DMSO-d6, 500 MHz, δ ppm): δ 0.80 (d, 6H, J = 5.6 Hz, CH(CH3)2), 1.42 (d, 3H, J = 6.0 Hz, CHCH3), 1.69–1.77 (m, 1H, CH(CH3)2), 2.35 (d, 2H, J = 6.7 Hz, Ph-CH2), 2.40–2.46 (m, 2H, pyrrolizine CH2-2), 2.97 (t, 2H, J = 6.6 Hz, pyrrolizine CH2-1), 3.89 (q, 1H, J = 7.0 Hz, CHCH3), 4.24 (t, 2H, J = 6.4 Hz, pyrrolizine CH2-3), 7.00 (d, 2H, J = 6.9 Hz, Ph (B) CH-3+CH-5), 7.11 (t, 2H, J = 7.9 Hz, Ph (A) CH-3+CH-5), 7.28 (d, 2H, J = 6.9 Hz, Ph (B) CH-2+CH-6), 7.38–7.41 (m, 2H, Ph (A) CH-2+CH-6), 9.43 (s, 1H, CONHCH), 10.20 (s, 1H, CONHPh). 13C-NMR (DMSO-d6, 125 MHz, δ ppm): δ 18.88, 22.60, 24.80, 25.66, 30.03, 44.67, 45.27, 49.70, 84.65, 114.92, 115.71 (d, J = 22.3 Hz, Ph (A) CH-3+CH-5), 118.65, 121.64 (d, J = 8.2 Hz, Ph (A) CH-2+CH-6), 126.78, 127.52, 129.40, 135.01 (d, J = 2.3 Hz, Ph (A) C-1), 138.63, 140.09, 146.43, 157.59, 158.69 (d, J = 240.2 Hz, Ph (A) C-4), 175.06. LC-MS: m/z 471.10 [M-H]+. Anal. Calcd. for C28H29FN4O2 (472.55): C, 71.17; H, 6.19; N, 11.86. Found C, 70.78; H, 5.81; N, 11.65.
(R,S)-6-(2-(3-Benzoylphenyl)propanamido)-7-cyano-N-(4-fluorophenyl)-2,3-dihydro-1H-pyrrolizine-5-carboxamide (8d) The title compound was prepared from the reaction of compound 5b (0.85 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ketoprofen (1.45 g, 5.9 mmol) according to the general procedure A. Compound 8d was obtained as white solid product, m.p. 240–2 °C, yield 59%. IRυmax/cm−1 3355, 3210 (NHs), 3025, 2996, 2879 (C-H aliphatic), 2219 (CN), 1663, 1634 (C=Os), 1616, 1564, 1505 (C=C, C=N), 1442, 1322, 1208 (C-N, C-O). 1H-NMR (DMSO-d6, 500 MHz, δ ppm): δ 1.47 (d, 3H, J = 6.0 Hz, CHCH3), 2.41–2.46 (m, 2H, pyrrolizine CH2-2), 2.97 (t, 2H, J = 6.7 Hz, pyrrolizine CH2-1), 4.05 (q, 1H, J = 6.8 Hz, CHCH3), 4.24 (t, 2H, J = 6.7 Hz, pyrrolizine CH2-3), 7.06 (t, 2H, J = 8.0 Hz, aromatic Hs), 7.31–7.34 (m, 2H, aromatic Hs), 7.46 (t, 1H, J = 7.2 Hz, aromatic H), 7.51–7.56 (m, 3H, aromatic Hs), 7.64–7.70 (m, 4H, aromatic Hs), 7.71 (s, 1H, Ph (B) CH-2). 9.35 (s, 1H, CONHCH), 10.23 (s, 1H, CONHPh). 13C-NMR (DMSO-d6, 125 MHz, δ ppm): δ 18.95, 24.80, 25.66, 45.45, 49.69, 84.51, 114.84, 115.74 (d, J = 22.3 Hz, Ph (A) CH-3+CH-5), 118.54, 121.55 (d, J = 7.9 Hz, Ph (A) CH-2+CH-6), 126.67, 128.93, 128.99, 129.06, 129.20, 130.06, 132.13, 133.20, 134.98 (d, J = 2.3 Hz, Ph (A) C-1), 137.39, 137.55, 141.91, 146.44, 157.61, 158.65 (d, J = 240.6 Hz, Ph (A) C-4), 174.38, 196.09. DEPT C135 (DMSO-d6, 125 MHz, δ ppm): δ 18.95, 24.80, 25.66, 45.45, 49.69, 115.74 (d, J = 22.3 Hz, Ph (A) CH-3+CH-5), 121.55 (d, J = 7.9 Hz, Ph (A) CH-2+CH-6), 128.93, 128.99, 129.06, 129.20, 130.06, 132.13, 133.20. LC-MS: m/z 519.10 [M-H]+. Anal. Calcd. for C31H25FN4O3 (520.55): C, 71.53; H, 4.84; N, 10.76. Found C, 71.32; H, 4.46; N, 11.23.
(R,S)-6-(2-(3-Benzoylphenyl)propanamido)-N-(4-bromophenyl)-7-cyano-2,3-dihydro-1H-pyrrolizine-5-carboxamide (8f)The title compound was prepared from the reaction of compound 5c (1.04 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ketoprofen (1.45 g, 5.9 mmol) according to the general procedure A. Compound 8f was obtained as white solid product, m.p. 229–31 °C, yield 61%. IRυmax/cm−1 3401, 3270 (NHs), 3063, 2965, 2874 (C-H aliphatic), 2222 (CN), 1659 (C=O), 1593, 1520 (C=C, C=N), 1423, 1315, 1270 (C-N, C-O). 1H-NMR (DMSO-d6, 500 MHz, δ ppm): δ 1.48 (d, 3H, J = 6.9 Hz, CHCH3), 2.44 (m, 2H, pyrrolizine CH2-2), 2.98 (t, 2H, J = 7.4 Hz, pyrrolizine CH2-1), 4.05 (q, 1H, J = 6.8 Hz, CHCH3), 4.25 (t, 2H, J = 7.1 Hz, pyrrolizine CH2-3), 7.29 (d, 2H, J = 8.5 Hz, aromatic Hs), 7.40 (d, 2H, J = 8.5 Hz, aromatic Hs), 7.47 (t, 1H, J = 7.6 Hz, aromatic H), 7.52–7.56 (m, 3H, aromatic Hs), 7.65–7.71 (m, 4H, aromatic Hs), 7.78 (s, 1H, Ph (B) CH-2). 9.43 (s, 1H, CONHCH), 10.24 (s, 1H, CONHPh). 13C-NMR (DMSO-d6, 125 MHz, δ ppm): δ 18.98, 24.82, 25.65, 45.46, 49.72, 84.57, 114.79, 115.71, 118.43, 121.60, 126.93, 128.96, 129.00, 129.05, 129.19, 130.04, 131.94, 132.15, 133.17, 137.42, 137.54, 138.04, 141.93, 146.59, 157.73. 174.36, 196.06. DEPT C135 (DMSO-d6, 125 MHz, δ ppm): δ 18.98, 24.82, 25.66, 45.46, 49.72, 121.60, 129.01, 129.19, 130.05, 131.94, 132.15, 133.17. LC-MS: m/z 581.20 [M+H]+. Anal. Calcd. for C31H25BrN4O3 (581.46): C, 64.03; H, 4.33; N, 9.64. Found C, 63.85; H, 4.69; N, 10.09.
(R,S)-N-(4-Chlorophenyl)-1-cyano-2-(2-(4-isobutylphenyl)propanamido)-5,6,7,8-tetrahydroindolizine-3-carboxamide (8g) The title compound was prepared from the reaction of compound 5d (0.94 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ibuprofen (1.22 g, 5.9 mmol) according to the general procedure A. Compound 8c was obtained as white solid product, m.p. 221–2 °C, yield 63%. IRυmax/cm−1 3347, 3282 (NHs), 3059, 2952, 2907 (C-H aliphatic), 2223 (CN), 1655 (C=Os), 1603, 1571 (C=C, C=N), 1492, 1377 (C-N, C-O). 1H-NMR (DMSO-d6, 500 MHz, δ ppm): δ 0.81 (d, 6H, J = 4.7 Hz, CH(CH3)2), 1.39 (d, 3H, J = 7.0 Hz, CHCH3), 1.70–1.75 (m, 1H, CH(CH3)2), 1.78–1.81 (m, 2H, indolizine CH2-7), 1.87–1.90 (m, 2H, indolizine CH2-6), 2.34 (d, 2H, J = 7.0 Hz, Ph-CH2), 2.83 (t, 2H, J = 5.7 Hz, indolizine CH2-8), 3.85 (q, 1H, J = 7.2 Hz, CHCH3), 4.10 (t, 2H, J = 5.3 Hz, indolizine CH2-5), 6.95 (d, 2H, J = 7.9 Hz, Ph (B) CH-3+CH-5), 7.23 (d, 2H, J = 7.9 Hz, Ph (B) CH-2+CH-6), 7.31 (d, 2H, J = 8.7 Hz, Ph (A) CH-3+CH-5), 7.43 (d, 2H, J =8.7 Hz, Ph (A) CH-2+CH-6), 9.73 (s, 1H, CONHCH), 10.01 (s, 1H, CONHPh). 13C-NMR (DMSO, 125 MHz, δ ppm): δ 18.86, 18.94, 22.57, 22.60, 22.63, 30.01, 44.70, 45.20, 45.64, 88.57, 114.70, 121.29, 121.79, 124.00, 127.46, 127.68, 128.99, 129.30, 137.76, 138.53, 139.99, 140.55, 157.86, 175.04. DEPT C135 (DMSO, 125 MHz, δ ppm): δ 18.86, 18.94, 22.57, 22.60, 22.63, 30.01, 44.70, 45.20, 45.64, 121.29, 127.46, 128.99, 129.30. LC-MS: m/z 501.00 [M-H]+. Anal. Calcd. for C29H31ClN4O2 (503.04): C, 69.24; H, 6.21; Cl, 7.05; N, 11.14. Found C, 69.39; H, 5.76; N, 11.42.
(R,S)-2-(2-(3-Benzoylphenyl)propanamido)-N-(4-chlorophenyl)-1-cyano-5,6,7,8-tetrahydroindolizine-3-carboxamide (8h) The title compound was prepared from the reaction of compound 5d (0.94 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ketoprofen (1.45 g, 5.9 mmol) according to the general procedure A. Compound 8h was obtained as white solid product, m.p. 252–4 °C, yield 55%. IRυmax/cm−1 3252, 3221 (NHs), 3084, 2974, 2959 (C-H aliphatic), 2217 (CN), 1662 (C=O), 1607, 1574, 1552 (C=C, C=N), 1489, 1314, 1259 (C-N, C-O). 1H-NMR (DMSO-d6, 500 MHz, δ ppm): δ 1.44 (d, 3H, J = 7.0 Hz, CHCH3), 1.76-1.81 (m, 2H, indolizine CH2-7), 1.86-1.91 (m, 2H, indolizine CH2-6), 2.82 (t, 2H, J = 6.0 Hz, indolizine CH2-8), 4.01 (q, 1H, J = 6.8 Hz, CHCH3), 4.10 (t, 2H, J = 5.5 Hz, indolizine CH2-5), 7.26 (d, 2H, J = 8.5 Hz, aromatic Hs), 7.38 (d, 2H, J = 8.5 Hz, aromatic Hs), 7.40–7.55 (m, 4H, aromatic Hs), 7.65–7.69 (m, 4H, aromatic Hs), 7.74 (s, 1H, Ph (B) CH-2), 9.70 (s, 1H, CONHCH), 10.11 (s, 1H, CONHPh). 13C-NMR (DMSO-d6, 125 MHz, δ ppm): δ 18.94, 22.56, 22.59, 45.36, 45.63, 52.92, 88.38, 114.65, 121.22, 123.94, 127.45, 128.85, 129.00, 129.09, 129.28, 130.05, 131.31, 132.09, 133.15, 137.45, 137.77, 139.35, 140.50, 141.85, 147.91, 164.53, 174.24, 196.05. DEPT C135 (DMSO-d6, 125 MHz, δ ppm): δ 18.93, 22.56, 22.59, 45.36, 45.63, 52.92, 121.22, 127.45, 128.85, 129.00, 129.10, 129.28, 130.06, 132.09, 133.15. LC-MS: m/z 549.20 [M-H]+. Anal. Calcd. for C32H27ClN4O3 (551.03): C, 69.75; H, 4.94; N, 10.17. Found C, 69.31; H, 4.67; N, 10.61.
(R,S)-N-(4-Bromophenyl)-1-cyano-2-(2-(4-isobutylphenyl)propanamido)-5,6,7,8-tetrahydroindolizine-3-carboxamide (8i) The title compound was prepared from the reaction of compound 5e (1.08 g, 3 mmol) with the acid chloride obtained from the reaction of thionyl chloride and ibuprofen (1.22 g, 5.9 mmol) according to the general procedure A. Compound 8i was obtained as white solid product, m.p. 235–7 °C, yield 54%. IRυmax/cm−1 3346, 3280 (NHs), 3056, 2952 (C-H aliphatic), 2222 (CN), 1656 (C=O), 1602, 1571, 1539 (C=C, C=N), 1489, 1310 (C-N, C-O). 1H-NMR (CDCl3, 500 MHz, δ ppm): δ 0.90 (d, 6H, J = 3.4 Hz, CH(CH3)2), 1.64 (d, 3H, J = 6.7 Hz, CHCH3), 1.80–1.88 (m, 3H, indolizine CH2-7+CH(CH3)2), 1.95–1.99 (m, 2H, indolizine CH2-6), 2.44 (d, 2H, J = 6.7 Hz, Ph-CH2), 2.86 (t, 2H, J = 6.1 Hz, indolizine CH2-8), 3.82 (q, 1H, J = 6.7 Hz, CHCH3), 4.14–4.27 (m, 2H, indolizine CH2-5), 7.05 (d, 2H, J = 7.0 Hz, Ph (B) CH-3+CH-5), 7.20 (d, 2H, J = 8.1 Hz, Ph (B) CH-2+CH-6), 7.28–7.31 (m, 2H, aromatic Hs), 7.38–7.41 (m, 3H, CONH+aromatic Hs), 9.67 (s, 1H, CONH). 13C-NMR (DMSO, 125 MHz, δ ppm): δ 18.09, 18.91, 22.38, 22.67, 22.73, 30.14, 44.97, 45.75, 46.81, 88.63, 113.75, 116.79, 121.01, 121.85, 123.45, 127.38, 129.97, 131.86, 136.40, 137.10, 140.49, 141.62, 157.55, 177.46. DEPT C135 (DMSO, 125 MHz, δ ppm): δ 18.09, 18.91, 22.38, 22.67, 22.74, 30.14, 44.97, 45.75, 46.81, 121.02, 127.38, 129.97, 131.86. LC-MS: m/z 547.00 [M+H]+. Anal. Calcd. for C29H31BrN4O2 (547.49): C, 63.62; H, 5.71; N, 10.23. Found C, 63.54; H, 5.46; N, 10.33.

4.2. Biological Evaluation

4.2.1. Antiproliferative Activity

Cell Culture

In this study, three cancer cell lines, MCF-7, A549, and HT-29 of the American Type Culture Collection (ATCC) were used in the evaluation of the antiproliferative activities of the new compounds. The cancer cells were cultured following the previously reported conditions [37].

Screening Assay

To evaluate the antiproliferative activities of the new compounds and their parent compounds, a quick screening MTT assay was used [37]. The cancer cell lines were treated with the hybrids 8a–i, and the parent compounds, ibuprofen, ketoprofen and compounds 5a–e at 5 μM. After 72 h of treatment, the absorbance of the purple formazan was determined using multi-plate reader at 570 nm and the growth% of the treated cells was determined and compared with the control.

MTT Assay

The antiproliferative activity of compounds 8a,e,f was evaluated against MCF-7 cells using MTT assay [38]. First, 96-well plates were used for the seeding of the cancer cells for 24 h. The cancer cells were treated test compounds 8a,e,f, and doxorubicin at different concentrations (0 µM–50 µM) for 72 h. The antiproliferative activity expressed as IC50 values for each of the four compounds were determined.

4.2.2. Cell Cycle Analysis

The effect of compounds 8a,e,f on cell cycle perturbation of MCF-7 cells was evaluated using flowcytometry (BD FAC SCalibur flow cytometer, BD Biosciences, Franklin Lakes, NJ, USA). The cancer cell was cultured into 6 well plates, then were treated with each of the test compounds at 5 μM for 48 h. The assay was conducted following the previous report [36].

4.2.3. Annexin V-FITC/PI Apoptosis Assay

Annexin V-FITC/PI double-staining assay was used to investigate the effect of compounds 8a,e,f on apoptotic events in MCF-7 cells compared to the control. In 6 well plates, the cancer cells were seeded overnight. The cancer cells were treated with 8a,e,f at 5 μM for 48 h. The assay was conducted according to our previous report [36]. NovoCyte flow cytometer (BD FAC SCalibur flow cytometer, BD Biosciences, Franklin Lakes, NJ, USA) was used to analyse the cancer cells with early/late apoptotic and necrotic changes.

4.3. Computational Studies

4.3.1. Molecular Docking Studies

Compounds (8a,e,f) were evaluated in a comparative molecular docking study into COX-1 (pdb code: 1EQG) [43] and COX-2 (pdb code: 1CX2) [44]. In addition, the parent drugs of the three hybrids 5a,c were also docked into the two COXs. The crystal structures of the two enzymes were obtained from protein data bank (http://www.rcsb.org/pdb) (accessed on 28 July 2021). AutoDock 4.2 [45] was used to dock the test compounds into the active sites of the two COXs. The study was performed following the previous report [46]. The generated 2/3D figures of the binding modes of the test compounds were created by discovery studio visualizer [47].

4.3.2. Drug-Likeness and ADME Studies

The molecular properties related to the pharmacokinetic/drug-likeness parameters of compounds 8a–i were evaluated by SwissADME (http://www.swissadme.ch/) (accessed on 24 September 2021) [50] and Molsoft (http://molsoft.com/mprop/) (accessed on 7 August 2021) webservers in the calculation of the physicochemical properties. The compounds were either sketched directly or imputed as SMILES. The detailed results are provided in the Supplementary data, Tables S1–S18.

Supplementary Materials

The following are available online. Supplementary data including copies of spectral data (IR, 1H-NMR, 13C-NMR DEPT C135 spectra) of compounds 8a–i are provided in the supplementary material (Figures S1–S82).

Author Contributions

Conceptualization, M.A.S.A., A.M.A., F.A.A., D.M.Z., A.N.A., A.M.G. and E.A.M.B.; methodology, M.A.S.A., A.M.A., F.A.A., D.M.Z., A.N.A., A.M.G. and E.A.M.B.; software, M.A.S.A., A.M.A., F.A.A., D.M.Z., A.N.A., A.M.G. and E.A.M.B.; validation, M.A.S.A., A.M.A., F.A.A., D.M.Z., A.N.A., A.M.G. and E.A.M.B.; formal analysis, M.A.S.A., A.M.A., F.A.A., D.M.Z., A.N.A., A.M.G. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to thank the Deanship of Scientific Research at Umm Al-Qura University, Makkah, KSA for supporting this work by Grant: 19-MED-1-01-0024.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data supporting the reported results are avilable in the Supplementary data File.

Acknowledgments

The authors would like to thank the Deanship of Scientific Research at Umm Al-Qura University for supporting this work by Grant: 19-MED-1-01-0024.

Conflicts of Interest

Authors declared that there is no conflict of interest and have approved the article.

Sample Availability

Samples of the compounds are available upon authors.

References

  1. Sun, J.; Wei, Q.; Zhou, Y.; Wang, J.; Liu, Q.H.; Xu, H. A systematic analysis of FDA-approved anticancer drugs. BMC Syst. Biol. 2017, 11, 87. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  3. Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ramos, A.; Sadeghi, S.; Tabatabaeian, H. Battling Chemoresistance in Cancer: Root Causes and Strategies to Uproot Them. Int. J. Mol. Sci. 2021, 22, 9451. [Google Scholar] [CrossRef]
  5. Damiana, T.S.T.; Dalm, S.U. Combination Therapy, a Promising Approach to Enhance the Efficacy of Radionuclide and Targeted Radionuclide Therapy of Prostate and Breast Cancer. Pharmaceutics 2021, 13, 674. [Google Scholar] [CrossRef] [PubMed]
  6. Smith, K.S.; Folz, B.A.; Adams, E.G.; Bhuyan, B.K. Synergistic and additive combinations of several antitumor drugs and other agents with the potent alkylating agent adozelesin. Cancer Chemother. Pharmacol. 1995, 35, 471–482. [Google Scholar] [CrossRef]
  7. Palmer, A.C.; Sorger, P.K. Combination Cancer Therapy Can Confer Benefit via Patient-to-Patient Variability without Drug Additivity or Synergy. Cell 2017, 171, 1678–1691.e13. [Google Scholar] [CrossRef] [Green Version]
  8. Carrick, S.; Parker, S.; Thornton, C.E.; Ghersi, D.; Simes, J.; Wilcken, N. Single agent versus combination chemotherapy for metastatic breast cancer. Cochrane Database Syst. Rev. 2009, 2009, CD003372. [Google Scholar] [CrossRef]
  9. Boshuizen, J.; Peeper, D.S. Rational Cancer Treatment Combinations: An Urgent Clinical Need. Mol. Cell 2020, 78, 1002–1018. [Google Scholar] [CrossRef]
  10. Delbaldo, C.; Michiels, S.; Syz, N.; Soria, J.-C.; le Chevalier, T.; Pignon, J.-P. Benefits of adding a drug to a single-agent or a 2-agent chemotherapy regimen in advanced non-small-cell lung cancer: A meta-analysis. JAMA 2004, 292, 470–484. [Google Scholar] [CrossRef]
  11. Riechelmann, R.P.; Tannock, I.F.; Wang, L.; Saad, E.D.; Taback, N.A.; Krzyzanowska, M.K. Potential drug interactions and duplicate prescriptions among cancer patients. J. Natl. Cancer Inst. 2007, 99, 592–600. [Google Scholar] [CrossRef]
  12. Szumilak, M.; Wiktorowska-Owczarek, A.; Stanczak, A. Hybrid Drugs—A Strategy for Overcoming Anticancer Drug Resistance? Molecules 2021, 26, 2601. [Google Scholar] [CrossRef]
  13. Hasan, M.; Leak, R.K.; Stratford, R.E.; Zlotos, D.P.; Witt-Enderby, P.A. Drug conjugates—An emerging approach to treat breast cancer. Pharmacol. Res. Perspect. 2018, 6, e00417. [Google Scholar] [CrossRef] [PubMed]
  14. Li, C.; Wallace, S. Polymer-drug conjugates: Recent development in clinical oncology. Adv. Drug Deliv. Rev. 2008, 60, 886–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Marshall, D.J.; Harried, S.S.; Murphy, J.L.; Hall, C.A.; Shekhani, M.S.; Pain, C.; Lyons, C.A.; Chillemi, A.; Malavasi, F.; Pearce, H.L.; et al. Extracellular Antibody Drug Conjugates Exploiting the Proximity of Two Proteins. Mol. Ther. 2016, 24, 1760–1770. [Google Scholar] [CrossRef] [Green Version]
  16. Donaghy, H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs 2016, 8, 659–671. [Google Scholar] [CrossRef]
  17. Mercadante, S. The use of anti-inflammatory drugs in cancer pain. Cancer Treat. Rev. 2001, 27, 51–61. [Google Scholar] [CrossRef]
  18. Vardy, J.; Agar, M. Nonopioid drugs in the treatment of cancer pain. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2014, 32, 1677–1690. [Google Scholar] [CrossRef] [Green Version]
  19. Hamoya, T.; Fujii, G.; Miyamoto, S.; Takahashi, M.; Totsuka, Y.; Wakabayashi, K.; Toshima, J.; Mutoh, M. Effects of NSAIDs on the risk factors of colorectal cancer: A mini review. Genes Environ. Off. J. Jpn. Environ. Mutagen Soc. 2016, 38, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Moris, D.; Kontos, M.; Spartalis, E.; Fentiman, I.S. The Role of NSAIDs in Breast Cancer Prevention and Relapse: Current Evidence and Future Perspectives. Breast Care 2016, 11, 339–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Plano, D.; Karelia, D.N.; Pandey, M.K.; Spallholz, J.E.; Amin, S.; Sharma, A.K. Design, Synthesis, and Biological Evaluation of Novel Selenium (Se-NSAID) Molecules as Anticancer Agents. J. Med. Chem. 2016, 59, 1946–1959. [Google Scholar] [CrossRef] [PubMed]
  22. Weninger, A.; Baecker, D.; Obermoser, V.; Egger, D.; Wurst, K.; Gust, R. Synthesis and Biological Evaluation of Zeise’s Salt Derivatives with Acetylsalicylic Acid Substructure. Int. J. Mol. Sci. 2018, 19, 1612. [Google Scholar] [CrossRef] [Green Version]
  23. Baecker, D.; Obermoser, V.; Kirchner, E.A.; Hupfauf, A.; Kircher, B.; Gust, R. Fluorination as tool to improve bioanalytical sensitivity and COX-2-selective antitumor activity of cobalt alkyne complexes. Dalton Trans. 2019, 48, 15856–15868. [Google Scholar] [CrossRef]
  24. Banekovich, C.; Ott, I.; Koch, T.; Matuszczak, B.; Gust, R. Synthesis and biological activities of novel dexibuprofen tetraacetylriboflavin conjugates. Bioorg. Med. Chem. Lett. 2007, 17, 683–687. [Google Scholar] [CrossRef]
  25. Curci, A.; Denora, N.; Iacobazzi, R.M.; Ditaranto, N.; Hoeschele, J.D.; Margiotta, N.; Natile, G. Synthesis, characterization, and in vitro cytotoxicity of a Kiteplatin-Ibuprofen Pt(IV) prodrug. Inorg. Chim. Acta 2018, 472, 221–228. [Google Scholar] [CrossRef]
  26. Zhang, L.; Liu, L.; Zheng, C.; Wang, Y.; Nie, X.; Shi, D.; Chen, Y.; Wei, G.; Wang, J. Synthesis and biological evaluation of novel podophyllotoxin-NSAIDs conjugates as multifunctional anti-MDR agents against resistant human hepatocellular carcinoma Bel-7402/5-FU cells. Eur. J. Med. Chem. 2017, 131, 81–91. [Google Scholar] [CrossRef]
  27. Garrido, M.; González-Arenas, A.; Camacho-Arroyo, I.; Cabeza, M.; Alcaraz, B.; Bratoeff, E. Effect of new hybrids based on 5,16-pregnadiene scaffold linked to an anti-inflammatory drug on the growth of a human astrocytoma cell line (U373). Eur. J. Med. Chem. 2015, 93, 135–141. [Google Scholar] [CrossRef] [PubMed]
  28. Gouda, A.M.; Ali, H.I.; Almalki, W.H.; Azim, M.A.; Abourehab, M.A.S.; Abdelazeem, A.H. Design, synthesis, and biological evaluation of some novel pyrrolizine derivatives as COX inhibitors with anti-inflammatory/analgesic activities and low ulcerogenic liability. Molecules 2016, 21, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Attalah, K.M.; Abdalla, A.N.; Aslam, A.; Ahmed, M.; Abourehab, M.A.S.; ElSawy, N.A.; Gouda, A.M. Ethyl benzoate bearing pyrrolizine/indolizine moieties: Design, synthesis and biological evaluation of anti-inflammatory and cytotoxic activities. Bioorg. Chem. 2020, 94, 103371. [Google Scholar] [CrossRef] [PubMed]
  30. Gouda, A.M.; Beshr, E.A.; Almalki, F.A.; Halawah, H.H.; Taj, B.F.; Alnafaei, A.F.; Alharazi, R.S.; Kazi, W.M.; AlMatrafi, M.M. Arylpropionic acid-derived NSAIDs: New insights on derivatization, anticancer activity and potential mechanism of action. Bioorg. Chem. 2019, 92, 103224. [Google Scholar] [CrossRef] [PubMed]
  31. Dhakane, V.D.; Chavan, H.V.; Thakare, V.N.; Adsul, L.K.; Shringare, S.N.; Bandgar, B.P. Novel ibuprofen prodrugs with improved pharmacokinetics and non-ulcerogenic potential. Med. Chem. Res. 2014, 23, 503–517. [Google Scholar] [CrossRef]
  32. Shanbhag, V.R.; Crider, A.M.; Gokhale, R.; Harpalani, A.; Dick, R.M. Ester and amide prodrugs of ibuprofen and naproxen: Synthesis, anti-inflammatory activity, and gastrointestinal toxicity. J. Pharm. Sci. 1992, 81, 149–154. [Google Scholar] [CrossRef] [PubMed]
  33. Marjanović, M.; Zorc, B.; Pejnović, L.; Zovko, M.; Kralj, M. Fenoprofen and Ketoprofen Amides as Potential Antitumor Agents. Chem. Biol. Drug Des. 2007, 69, 222–226. [Google Scholar] [CrossRef] [PubMed]
  34. Almalki, F.A.; Abdalla, A.N.; Shawky, A.M.; el Hassab, M.A.; Gouda, A.M. In Silico Approach Using Free Software to Optimize the Antiproliferative Activity and Predict the Potential Mechanism of Action of Pyrrolizine-Based Schiff Bases. Molecules 2021, 26, 4002. [Google Scholar] [CrossRef]
  35. Shawky, A.M.; Abdalla, A.N.; Ibrahim, N.A.; Abourehab, M.A.S.; Gouda, A.M. Discovery of new pyrimidopyrrolizine/indolizine-based derivatives as P-glycoprotein inhibitors: Design, synthesis, cytotoxicity, and MDR reversal activities. Eur. J. Med. Chem. 2021, 218, 113403. [Google Scholar] [CrossRef]
  36. Shawky, A.M.; Ibrahim, N.A.; Abdalla, A.N.; Abourehab, M.A.S.; Gouda, A.M. Novel pyrrolizines bearing 3,4,5-trimethoxyphenyl moiety: Design, synthesis, molecular docking, and biological evaluation as potential multi-target cytotoxic agents. J. Enzym. Inhib. Med. Chem. 2021, 36, 1313–1333. [Google Scholar] [CrossRef] [PubMed]
  37. Shawky, A.M.; Abourehab, M.A.S.; Abdalla, A.N.; Gouda, A.M. Optimization of pyrrolizine-based Schiff bases with 4-thiazolidinone motif: Design, synthesis and investigation of cytotoxicity and anti-inflammatory potency. Eur. J. Med. Chem. 2020, 185, 111780. [Google Scholar] [CrossRef] [PubMed]
  38. Shawky, A.M.; Ibrahim, N.A.; Abourehab, M.A.S.; Abdalla, A.N.; Gouda, A.M. Pharmacophore-based virtual screening, synthesis, biological evaluation, and molecular docking study of novel pyrrolizines bearing urea/thiourea moieties with potential cytotoxicity and CDK inhibitory activities. J. Enzym. Inhib. Med. Chem. 2021, 36, 15–33. [Google Scholar] [CrossRef]
  39. Amujuri, D.; Siva, B.; Poornima, B.; Sirisha, K.; Sarma, A.V.S.; Lakshma Nayak, V.; Tiwari, A.K.; Purushotham, U.; Suresh Babu, K. Synthesis and biological evaluation of Schizandrin derivatives as potential anti-cancer agents. Eur. J. Med. Chem. 2018, 149, 182–192. [Google Scholar] [CrossRef]
  40. Mazaleuskaya, L.L.; Theken, K.N.; Gong, L.; Thorn, C.F.; FitzGerald, G.A.; Altman, R.B.; Klein, T.E. PharmGKB summary: Ibuprofen pathways. Pharmacogenet. Genom. 2015, 25, 96–106. [Google Scholar] [CrossRef] [Green Version]
  41. Alkatheeri, N.A.; Wasfi, I.A.; Lambert, M. Pharmacokinetics and metabolism of ketoprofen after intravenous and intramuscular administration in camels. J. Vet. Pharmacol. Ther. 1999, 22, 127–135. [Google Scholar] [CrossRef]
  42. Qandil, A.M. Prodrugs of nonsteroidal anti-inflammatory drugs (NSAIDs), more than meets the eye: A critical review. Int. J. Mol. Sci. 2012, 13, 17244–17274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Selinsky, B.S.; Gupta, K.; Sharkey, C.T.; Loll, P.J. Structural analysis of NSAID binding by prostaglandin H2 synthase: Time-dependent and time-independent inhibitors elicit identical enzyme conformations. Biochemistry 2001, 40, 5172–5180. [Google Scholar] [CrossRef]
  44. Kurumbail, R.G.; Stevens, A.M.; Gierse, J.K.; McDonald, J.J.; Stegeman, R.A.; Pak, J.Y.; Gildehaus, D.; Miyashiro, J.M.; Penning, T.D.; Seibert, K.; et al. Structural basis for selective inhibition of cyclooxygenase-2 by anti-inflammatory agents. Nature 1996, 384, 644–648. [Google Scholar] [CrossRef]
  45. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [Green Version]
  46. Mohassab, A.M.; Hassan, H.A.; Abdelhamid, D.; Gouda, A.M.; Gomaa, H.A.M.; Youssif, B.G.M.; Radwan, M.O.; Fujita, M.; Otsuka, M.; Abdel-Aziz, M. New quinoline/1,2,4-triazole hybrids as dual inhibitors of COX-2/5-LOX and inflammatory cytokines: Design, synthesis, and docking study. J. Mol. Struct. 2021, 1244, 130948. [Google Scholar] [CrossRef]
  47. Dassault Systems BIOVIA. Discovery Studio Visualizer, v16.1.0.15350; Dassault Systems: San Diego, CA, USA, 2016. [Google Scholar]
  48. Reichel, A.; Lienau, P. Pharmacokinetics in Drug Discovery: An Exposure-Centred Approach to Optimising and Predicting Drug Efficacy and Safety. Handb. Exp. Pharmacol. 2016, 232, 235–260. [Google Scholar] [CrossRef] [PubMed]
  49. Tuntland, T.; Ethell, B.; Kosaka, T.; Blasco, F.; Zang, R.X.; Jain, M.; Gould, T.; Hoffmaster, K. Implementation of pharmacokinetic and pharmacodynamic strategies in early research phases of drug discovery and development at Novartis Institute of Biomedical Research. Front. Pharmacol. 2014, 5, 174. [Google Scholar] [CrossRef] [Green Version]
  50. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 2001, 46, 3–26. [Google Scholar] [CrossRef]
  52. Zhao, Y.H.; Abraham, M.H.; Le, J.; Hersey, A.; Luscombe, C.N.; Beck, G.; Sherborne, B.; Cooper, I. Rate-Limited Steps of Human Oral Absorption and QSAR Studies. Pharm. Res. 2002, 19, 1446–1457. [Google Scholar] [CrossRef] [PubMed]
Figure 1. NSAIDs complexes, hybrids, and conjugates of with anticancer activities: I, Se-aspirin complex reported by [21]; IIa–c, Zeise’s Salt derivatives of acetylsalicylic acid [22]; III, cobalt alkyne derivatives of fluorinated acetylsalicylic acid [23]; IVa–e, riboflavin-dexibuprofen conjugates [24]; V, ibuprofen-kiteplatin Pt(IV) conjugate [25]; VI, ibuprofen-podophyllotoxin conjugate [26]; VII, 5,16-pregnadiene-ibuprofen hybrid [27].
Figure 1. NSAIDs complexes, hybrids, and conjugates of with anticancer activities: I, Se-aspirin complex reported by [21]; IIa–c, Zeise’s Salt derivatives of acetylsalicylic acid [22]; III, cobalt alkyne derivatives of fluorinated acetylsalicylic acid [23]; IVa–e, riboflavin-dexibuprofen conjugates [24]; V, ibuprofen-kiteplatin Pt(IV) conjugate [25]; VI, ibuprofen-podophyllotoxin conjugate [26]; VII, 5,16-pregnadiene-ibuprofen hybrid [27].
Molecules 26 06582 g001
Figure 2. Chemical structure of compounds VIIIa–c: VIIIa, a hybrid of ibuprofen with 6-amino-N-(4-bromophenyl)-7-cyano-2,3-dihydro-1H-pyrrolizine-5-carboxamide that exhibited in vivo anti-inflammatory activity [25]; VIIIb,c, ibuprofen-pyrrolizine/indolizine hybrids with cytotoxic activity [26].
Figure 2. Chemical structure of compounds VIIIa–c: VIIIa, a hybrid of ibuprofen with 6-amino-N-(4-bromophenyl)-7-cyano-2,3-dihydro-1H-pyrrolizine-5-carboxamide that exhibited in vivo anti-inflammatory activity [25]; VIIIb,c, ibuprofen-pyrrolizine/indolizine hybrids with cytotoxic activity [26].
Molecules 26 06582 g002
Figure 3. Rational design of the new hybrids. The new hybrids (scaffold B) were obtained by combining ibuprofen or ketoprofen with the pyrrolizine or indolizine derivatives (scaffold A).
Figure 3. Rational design of the new hybrids. The new hybrids (scaffold B) were obtained by combining ibuprofen or ketoprofen with the pyrrolizine or indolizine derivatives (scaffold A).
Molecules 26 06582 g003
Scheme 1. Reagents and conditions: (a) (CH3)2SO4, benzene, CH2(CN)2; (b) ClCH2COCl, g. acetic acid, CH2COONa, rt, 2 h; (c) acetone, K2CO3, reflux, 24 h; (d) SOCl2, heat, 2 h; (e) DCM, TEA, rt, stir, 24 h.
Scheme 1. Reagents and conditions: (a) (CH3)2SO4, benzene, CH2(CN)2; (b) ClCH2COCl, g. acetic acid, CH2COONa, rt, 2 h; (c) acetone, K2CO3, reflux, 24 h; (d) SOCl2, heat, 2 h; (e) DCM, TEA, rt, stir, 24 h.
Molecules 26 06582 sch001aMolecules 26 06582 sch001b
Figure 4. Anti-proliferative effect of the tested compounds on the viability of MCF-7 cells. Columns are mean of cell viability percent after treatment with compounds 5a-e, 8a-i, ibuprofen and ketoprofen at 5 μM for 72 h ±SD (n = 6). MTT cell viability assay experiment was conducted three times. p-value determined by two-tailed unpaired t-test. * indicates significant difference versus control at p < 0.05 (*), p < 0.01 (**). # indicates significant difference versus Ibuprofen at p < 0.05 (#), p < 0.01 (##). @ indicates significant difference versus Ketoprofen at p < 0.01 (@).
Figure 4. Anti-proliferative effect of the tested compounds on the viability of MCF-7 cells. Columns are mean of cell viability percent after treatment with compounds 5a-e, 8a-i, ibuprofen and ketoprofen at 5 μM for 72 h ±SD (n = 6). MTT cell viability assay experiment was conducted three times. p-value determined by two-tailed unpaired t-test. * indicates significant difference versus control at p < 0.05 (*), p < 0.01 (**). # indicates significant difference versus Ibuprofen at p < 0.05 (#), p < 0.01 (##). @ indicates significant difference versus Ketoprofen at p < 0.01 (@).
Molecules 26 06582 g004
Figure 5. Anti-proliferative effect of the tested compounds on the viability of A549 cells. Columns are mean of cell viability percent after treatment with compounds 5a–d, 8a–i, ibuprofen and ketoprofen at 5 μM for 72 h ± SD (n = 6). MTT cell viability assay experiment was conducted three times. p-value determined by two-tailed unpaired t-test. * indicates significant difference versus control at p < 0.05 (*), p < 0.01 (**). # indicates significant difference versus Ibuprofen at p < 0.05 (#), p < 0.01 (##). @ indicates significant difference versus Ketoprofen at p < 0.01 (@).
Figure 5. Anti-proliferative effect of the tested compounds on the viability of A549 cells. Columns are mean of cell viability percent after treatment with compounds 5a–d, 8a–i, ibuprofen and ketoprofen at 5 μM for 72 h ± SD (n = 6). MTT cell viability assay experiment was conducted three times. p-value determined by two-tailed unpaired t-test. * indicates significant difference versus control at p < 0.05 (*), p < 0.01 (**). # indicates significant difference versus Ibuprofen at p < 0.05 (#), p < 0.01 (##). @ indicates significant difference versus Ketoprofen at p < 0.01 (@).
Molecules 26 06582 g005
Figure 6. Anti-proliferative effect of the tested compounds on the viability of HT-29 cells. Columns are mean of cell viability percent after treatment with compounds 5a–d, 8a–i, ibuprofen and ketoprofen at 5 μM for 72 h ± SD (n = 6). MTT cell viability assay experiment was conducted three times. p-value determined by two-tailed unpaired t-test. * indicates significant difference versus control at p < 0.01 (**). # indicates significant difference versus Ibuprofen at p < 0.05 (#), p < 0.01 (##). @ indicates significant difference versus Ketoprofen at p < 0.05 (@), p < 0.01 (@@).
Figure 6. Anti-proliferative effect of the tested compounds on the viability of HT-29 cells. Columns are mean of cell viability percent after treatment with compounds 5a–d, 8a–i, ibuprofen and ketoprofen at 5 μM for 72 h ± SD (n = 6). MTT cell viability assay experiment was conducted three times. p-value determined by two-tailed unpaired t-test. * indicates significant difference versus control at p < 0.01 (**). # indicates significant difference versus Ibuprofen at p < 0.05 (#), p < 0.01 (##). @ indicates significant difference versus Ketoprofen at p < 0.05 (@), p < 0.01 (@@).
Molecules 26 06582 g006
Figure 7. Flow cytometry histograms showing the effect of compound 8a,e,f (5 μM) on cell cycle distribution of MCF-7 cells after 48 h treatment with (A) control; (B) 8a; (C) 8e; (D) 8f, (n = 3). Experiment was repeated 3×.
Figure 7. Flow cytometry histograms showing the effect of compound 8a,e,f (5 μM) on cell cycle distribution of MCF-7 cells after 48 h treatment with (A) control; (B) 8a; (C) 8e; (D) 8f, (n = 3). Experiment was repeated 3×.
Molecules 26 06582 g007
Figure 8. Evaluation of apoptosis-inducing activity of compounds 8a,e,f in MCF-7 cells using annexin V FITC/PI staining assay: (A) control; (B) 8a; (C) 8e; (D) 8f, (n = 3). x-axis: annexin V/FITC, y-axis: PI. C1: necrosis, PI+/annexin V−); C2: (late apoptosis, PI+/ annexin V+); C3: (living cells, PI−/annexin V−); C4: (early apoptosis, PI−/annexin V+).
Figure 8. Evaluation of apoptosis-inducing activity of compounds 8a,e,f in MCF-7 cells using annexin V FITC/PI staining assay: (A) control; (B) 8a; (C) 8e; (D) 8f, (n = 3). x-axis: annexin V/FITC, y-axis: PI. C1: necrosis, PI+/annexin V−); C2: (late apoptosis, PI+/ annexin V+); C3: (living cells, PI−/annexin V−); C4: (early apoptosis, PI−/annexin V+).
Molecules 26 06582 g008
Figure 9. Binding modes/interactions of best-fitting conformation of 8a (shown as stick, colored by element) into the active site of COX-1 (pdb code: 1EQG): (A) 3D binding mode of 8a superposed with ibuprofen (yellow stacks); (B) 2D binding mode of 8a into COX-1 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Figure 9. Binding modes/interactions of best-fitting conformation of 8a (shown as stick, colored by element) into the active site of COX-1 (pdb code: 1EQG): (A) 3D binding mode of 8a superposed with ibuprofen (yellow stacks); (B) 2D binding mode of 8a into COX-1 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Molecules 26 06582 g009
Figure 10. Binding modes/interactions of best-fitting conformation of 8a (shown as stick, colored by element) into the active site of COX-2 (pdb code: 1CX2): (A) 3D binding mode of 8a superposed with SC588 (yellow stacks); (B) 2D binding mode of 8a into COX-2 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Figure 10. Binding modes/interactions of best-fitting conformation of 8a (shown as stick, colored by element) into the active site of COX-2 (pdb code: 1CX2): (A) 3D binding mode of 8a superposed with SC588 (yellow stacks); (B) 2D binding mode of 8a into COX-2 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Molecules 26 06582 g010
Figure 11. Binding modes/interactions of best-fitting conformation of 8e (shown as stick, colored by element) into the active site of COX-1 (pdb code: 1EQG): (A) 3D binding mode of 8e superposed with ibuprofen (yellow stacks); (B) 2D binding mode of 8e into COX-1 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Figure 11. Binding modes/interactions of best-fitting conformation of 8e (shown as stick, colored by element) into the active site of COX-1 (pdb code: 1EQG): (A) 3D binding mode of 8e superposed with ibuprofen (yellow stacks); (B) 2D binding mode of 8e into COX-1 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Molecules 26 06582 g011
Figure 12. Binding modes/interactions of best-fitting conformation of 8e (shown as stick, colored by element) into the active site of COX-2 (pdb code: 1CX2): (A) 3D binding mode of 8e superposed with SC588 (yellow stacks); (B) 2D binding mode of 8e into COX-2 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Figure 12. Binding modes/interactions of best-fitting conformation of 8e (shown as stick, colored by element) into the active site of COX-2 (pdb code: 1CX2): (A) 3D binding mode of 8e superposed with SC588 (yellow stacks); (B) 2D binding mode of 8e into COX-2 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Molecules 26 06582 g012
Figure 13. Binding modes/interactions of best-fitting conformation of 8f (shown as stick, colored by element) into the active site of COX-1 (pdb code: 1EQG): (A) 3D binding mode of 8f superposed with ibuprofen (yellow stacks); (B) 2D binding mode of 8f into COX-1 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Figure 13. Binding modes/interactions of best-fitting conformation of 8f (shown as stick, colored by element) into the active site of COX-1 (pdb code: 1EQG): (A) 3D binding mode of 8f superposed with ibuprofen (yellow stacks); (B) 2D binding mode of 8f into COX-1 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Molecules 26 06582 g013
Figure 14. Binding modes/interactions of best-fitting conformation of 8f (shown as stick, colored by element) into the active site of COX-2 (pdb code: 1CX2): (A) 3D binding mode of 8f superposed with SC588 (yellow stacks); (B) 2D binding mode of 8f into COX-2 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Figure 14. Binding modes/interactions of best-fitting conformation of 8f (shown as stick, colored by element) into the active site of COX-2 (pdb code: 1CX2): (A) 3D binding mode of 8f superposed with SC588 (yellow stacks); (B) 2D binding mode of 8f into COX-2 showing different types of binding interactions, hydrogen atoms were omitted for clarity, the figure generated using Discovery Studio Visualizer (V16.1.0.15350).
Molecules 26 06582 g014
Table 1. IC50 values of compounds 8a,e,f and doxorubicin against MCF-7.
Table 1. IC50 values of compounds 8a,e,f and doxorubicin against MCF-7.
Comp. NoIC50 (µM)
8a7.61 ± 0.44
8e1.07 ± 0.06
8f3.16 ± 0.18
Doxorubicin2.07 ± 0.12
Results were presented as mean ± S.D. (n = 3), after 72 h treatment with test compounds or vehicle (control).
Table 2. Cell cycle analysis of MCF-7 cells treated with compounds 8a,e,f (48 h).
Table 2. Cell cycle analysis of MCF-7 cells treated with compounds 8a,e,f (48 h).
Comp. Cell Cycle Stage (% ± SD)Effect
Pre-G1G1SG2/M
Control1.01 ± 0.1056.11 ± 7.3440.22 ± 6.783.07 ± 1.07---
8a6.57 ± 1.8048.41 ± 4.3145.61 ± 4.331.00 ± 0.06Increase of cells at preG1 and S
8e10.12 ± 1.9040.22 ± 3.9949.02 ± 3.881.76 ± 0.32Increase of cells at preG1 and S
8f5.81 ± 0.3260.19 ± 8.8135.17 ± 3.510.64 ± 0.15Increase of cells at preG1 and G1
Cell cycle phases of MCF-7 cells (% ± SD) after treatment with the test compounds at 5 μM for 48 h. The experiment was repeated 3×.
Table 3. Annexin V-FITC/PI staining assay results in MCF-7 cells after treatment with 8a,e,f.
Table 3. Annexin V-FITC/PI staining assay results in MCF-7 cells after treatment with 8a,e,f.
Comp.Living
Cells
ApoptosisNecrosis
EarlyLate
Control 94.50 ± 3.000.60 ± 0.200.80 ± 0.204.10 ± 1.00
8a85.00 ± 3.0015.00 ± 3.200.00 ± 0.000.00 ± 0.00
8e69.20 ± 2.3030.30 ± 3.010.00 ± 0.000.00 ± 0.00
8f89.10 ± 2.0211.03 ± 1.110.00 ± 0.000.00 ± 0.00
MCF-7 cells were treated with the test compounds for 48 h, data shown are % mean ± SD (n = 3). Experiment was repeated 3×.
Table 4. Results of the docking of 8a,e,f into ovine COX-1/2 in comparison to the parents 5a,c and the co-crystallized ligands (ibuprofen/ SC-558).
Table 4. Results of the docking of 8a,e,f into ovine COX-1/2 in comparison to the parents 5a,c and the co-crystallized ligands (ibuprofen/ SC-558).
Target
(pdb)
Comp. NoΔGb aKi bHBs cAtoms in H-BondingLength d
(Å)
In LigandIn COX-1
Cox-1
(1EQG)
5a−8.07 1.21 μM4OCH3Agr1202.11
OCH3Tyr3552.23
NH2Tyr3852.08
COSer5301.87
5c−8.78366.84 nM2CNArg120 1.98
CNArg120 2.49
8a−10.36 25.61 nM5*COsAgr1201.70, 2.76
COsTyr3551.83, 2.88
OCH3Gly526*2.22
8e−10.52 19.38 nM-e---
8f−10.5518.44 nM2COVal116*2.28
COArg1202.02
Ibu.−8.43664 nM3C=O Arg1201.71
COOHArg1201.79
COOHTyr3551.83
COX-2
(1CX2)
5a−8.61489.47 nM4OCH3His901.95
OCH3Gln1922.66
NH2Met5222.51
NH2Val5232.01
5c−8.56530.05 nM2NH2Val5232.24, 2.28
8a−11.04 8.05 nM3CNArg120 2.60
COSer353*2.73, 3.04
8e−10.7014.42 nM6COHis90*2.05, 2.26
CONHLeu3521.98
CN, COArg5131.94, 2.73
BrGly5262.55
8f−12.56 618.28 pM2COCOArg513 *
Ser530 *
2.69
2.42
SC-588−10.7812.52 nM5CF3Arg120*2.32–2.44
CF3, Pyrazole NTyr3552.58, 3.06
a Binding free energy; b inhibition constant; c number of hydrogen bonds; d length in angstrom (Å), e no hydrogen bonds detected, * indicated the functional groups/amino acids involved in carbon hydrogen bonds; Ibu, ibuprofen; SC-588, 4-[5-(4-bromophenyl)-3-(trifluoromethyl)pyrazol-1-yl]benzenesulfonamide.
Table 5. Molecular properties related to drug-likeness of compounds 3, 8a–i, ibuprofen and ketoprofen.
Table 5. Molecular properties related to drug-likeness of compounds 3, 8a–i, ibuprofen and ketoprofen.
Comp. NoPhysicochemical PropertiesLipinski’s Rule%Abs aDLS b
MW MPSAMlogPRBsHAHD
8a484.59 70.29 2.641042Yes84.751.54
8b532.59 83.762.141052Yes c80.101.06
8c472.55 62.75 3.34942Yes87.351.76
8d520.55 76.21 2.84952Yes c82.711.29
8e533.46 62.75 3.53932Yes c87.351.59
8f581.46 76.21 3.03942Yes c82.711.13
8g503.04 62.38 3.63932Yes c87.482.03
8h551.03 75.85 3.12942Yes c82.831.57
8i547.49 62.38 3.73932Yes c87.481.73
VI602.67 82.91 3.711090Yes c80.401.01
Ibu206.28 28.65 3.13421Yes99.120.65
Ket254.28 42.11 2.69431Yes94.470.57
a %Abs = 109 − (0.345 × TPSA); MV, molecular volume (A3); b DLS, drug-likeness score; c one violation, molecular weight >500; MolPSA, and DLS were calculated using Molsoft (http://molsoft.com/mprop/) (accessed on 1 October 2021); Ibu, ibuprofen; Ket, ketoprofen.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abourehab, M.A.S.; Alqahtani, A.M.; Almalki, F.A.; Zaher, D.M.; Abdalla, A.N.; Gouda, A.M.; Beshr, E.A.M. Pyrrolizine/Indolizine-NSAID Hybrids: Design, Synthesis, Biological Evaluation, and Molecular Docking Studies. Molecules 2021, 26, 6582. https://doi.org/10.3390/molecules26216582

AMA Style

Abourehab MAS, Alqahtani AM, Almalki FA, Zaher DM, Abdalla AN, Gouda AM, Beshr EAM. Pyrrolizine/Indolizine-NSAID Hybrids: Design, Synthesis, Biological Evaluation, and Molecular Docking Studies. Molecules. 2021; 26(21):6582. https://doi.org/10.3390/molecules26216582

Chicago/Turabian Style

Abourehab, Mohammed A. S., Alaa M. Alqahtani, Faisal A. Almalki, Dana M. Zaher, Ashraf N. Abdalla, Ahmed M. Gouda, and Eman A. M. Beshr. 2021. "Pyrrolizine/Indolizine-NSAID Hybrids: Design, Synthesis, Biological Evaluation, and Molecular Docking Studies" Molecules 26, no. 21: 6582. https://doi.org/10.3390/molecules26216582

APA Style

Abourehab, M. A. S., Alqahtani, A. M., Almalki, F. A., Zaher, D. M., Abdalla, A. N., Gouda, A. M., & Beshr, E. A. M. (2021). Pyrrolizine/Indolizine-NSAID Hybrids: Design, Synthesis, Biological Evaluation, and Molecular Docking Studies. Molecules, 26(21), 6582. https://doi.org/10.3390/molecules26216582

Article Metrics

Back to TopTop