You are on page 1of 7

Journal of Molecular Structure 1264 (2022) 133234

Contents lists available at ScienceDirect

Journal of Molecular Structure


journal homepage: www.elsevier.com/locate/molstr

Synthesis, characterization, computational, anticancer and


anti-inflammatory studies of pregabalin esters and their complexes
with heavy metals
Ayesha Asbat a, Farooq Saleem a,∗, Saima Najm b, Javed Iqbal a, Muhammad Ali Syed a,
Sarfraz Ahmad c, Sana Hanif a, Muhammad Azeem a
a
Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
b
Faculty of Pharmacy, Lahore College of Pharmaceutical Sciences, Lahore, Pakistan
c
Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia

a r t i c l e i n f o a b s t r a c t

Article history: The current study entails a series of pregabalin ester complexed with zinc and copper; further character-
Received 22 June 2021 ized by spectroscopic techniques and biological assays. Docking analysis revealed three complexes with
Revised 17 February 2022
favorable binding interactions which were further subjected to in-vitro anti-inflammatory and anticancer
Accepted 4 May 2022
activities. The complex-5 displayed most potent anti-inflammatory activity against Luminol-enhanced
Available online 18 May 2022
Chemiluminescence assay with IC50 value of 7.7 ± 0.8 μM as compared to standard Ibuprofen with IC50
Keywords: value of 11.2 ± 1.9 μM; whereas none of them showed significant outcomes against HeLa cell lines for
Pregabalin antiproliferative activity as compared with standard doxorubicin. Computational studies were carried out
Esters using the reported crystal structure of multidrug resistant Alpha2S protein (PDB-ID: 6ND9), neurokinin-
Auto dock 1 receptor (PDB-ID: 2KSA) and dihydrofolate reductase (PDB-ID: 3GHW); indicating the compound 3
Luminol showed significant interactions at all receptors. As the analogues were more active than parent drug in
Antitumor
some respects; therefore, the expansion of the project can be planned to carry out more pharmacological
Anti-inflammatory
screening.
© 2022 Elsevier B.V. All rights reserved.

1. Introduction drowsiness while using it in generalized anxiety disorder. However,


it would be safe to use these types of medications at night and
Pregabalin, [3 - (amino methyl hexonic acid)] is used in the avoid them to use during traveling [8].
treatment of epilepsy [1]. It was synthesized in 1990 as an an- On the high-pitched side, many drugs possess improved
ticonvulsant drug [2]. The drug treats convulsions at the dose of toxicological and pharmacological properties (anti-inflammatory
600 mg/day. However, some patients feel dizziness at initial dose and anti-cancer) in the form of metal complexes. Some metal
[3]. Numerous scientists have used functional MRI to study brain complexes e.g. silver metal complex was synthesized for anti-
systems; most of them have used graph theory analysis. However, inflammatory activity and the quinolone derivatives along with
functional techniques can be analyzed by the effects of antiepilep- their antimony (III) complexes were assessed for their cytotoxicity
tic drugs [4]. The drug was accepted to treat partial seizures in against HL-60 and Jurkat human leukemia cell lines [9,10]. How-
adults as well as to cure neuropathic pain and diabetic neuropathy ever, some essential trace metals including zinc, copper and iron
[5]. Pregabalin (PGB) binds with high affinity to the R2-δ subunit enable our body to function normally and their deficiency leads
of voltage gated calcium channels in cell membrane. Nitroalkane- to abnormal growth, poor immunity and decline oxygen transport
mediated cycloproponation as a central step was used to prepare to body [11]. A new platinum (IV) complex, [Pt(Cl)2(Pregabalin)2],
some new amino acids [6,7]. PGB lacks plasma protein binding and holding the drug PGB was synthesized, characterized by differ-
so it has no interactions with other anticonvulsants, antidiabetics, ent techniques and observed to bind with Human serum albumin
and oral contraceptives. However, it readily infiltrates the blood- (HSA) and causes strong fluorescence quenching of HAS [11,12].
brain barrier [1]. PGB shows adverse effects such as dizziness and Besides, metal complexes of PGB derivatives have played a cen-
tral role in improvement of co-ordination chemistry. In this study,
various esters and metal complexes of PGB were synthesized. For

Corresponding author. better understanding between ligands and targets, molecular dock-
E-mail address: farooq.saleem@pharm.uol.edu.pk (F. Saleem).

https://doi.org/10.1016/j.molstruc.2022.133234
0022-2860/© 2022 Elsevier B.V. All rights reserved.
A. Asbat, F. Saleem, S. Najm et al. Journal of Molecular Structure 1264 (2022) 133234

Scheme 1. General scheme of synthesis of pregabalin derivatives and their metal complexes (Zn and Cu).
M = ZnCl2, Cu(CH3 COO)2. H2 O 1=pregabalin; 2=formaldehyde; 3=Ligand formaldehyde with PGB; 4 a, b = pregabalin ester with metal complexes (Zn and Cu).

ing studies was planned and assessed their interactions against α 2- 4.69 for NH2 group. All other peaks at 2.24, 2.86 and 4.69 showed
δ −1 subunit (PDB-ID: 6ND9); neurokinin-1 (PDB ID: 2KSA) and di- presence of -CH3 group. Resultantly, above mentioned peaks con-
hydrofolate reductase (PDB ID: 3GHW). The findings explored the firm the structure of compound 3. 1 HNMR data of compound 4a
significant anticancer and anti-inflammatory potential PGB deriva- showed singlet peak at δ = 3.55 for -CH3 functional group and also
tives, making them suitable for advanced pharmacological screen- showed a clear singlet at δ = 4.70 which indicated the presence of
ing. –NH2 group and hence this spectroscopic analysis shows the con-
firmation of 4a. The absorption bands at 3266.8, 2963.9, 1669.8,
1546.8, 1485, 1388.4 and 1276.6 cm−1 confirmed the presence of
2. Results and discussion NH, -CH, -C=O, -NH2, -CH3,=CH2 and C-O, respectively so we have
seen the confirmation of compound 4b via this spectroscopic anal-
2.1. Chemistry ysis.

The target compounds were synthesized as sketched in


Scheme 1. The synthesis of PGB esters (3) were achieved by reac- 2.2. Solubility studies
tion of compound 1 with 2 (Scheme 1). The synthesis of PGB esters
is the main intermediate for synthesis of our target compounds. The solubility of newly synthesized metal complexes was eval-
Moreover, zinc chloride and copper acetate form coordinate bonds uated. A measured amount of each compound was dissolved in
with derivatives by using methanol to complete the formation weighed amount of solvent and clarity of solution was observed.
of required heavy metal complexes (4a and 4b) of PGB [12]. Fi- Water, methanol, ethanol, DMSO and chloroform were used for this
nally, the structure elucidation was achieved by FTIR, 1 H NMR and study. For good solubility the threshold value of <0.1/100 ml with
ESI-MS spectra. In FTIR spectra all compounds showed carbonyl the clear colored solution in a given solvent was considered [13].
stretching vibrations in the range of 1668–1670 cm−1 . Initially we The solubility of various ligands and their metal complexes is given
were expecting an open chain structure of coordinate complexes; in Table S2.
however, characterization confirmed the presence of straight chain
structures of amines (see supplementary Figs. S1,S2). FTIR spectra 2.3. Computational studies
of PGB ester and its metal complexes proved the straight chain
structure of amines. However, in the 1 H NMR spectra the multi- Molecular docking studies were used to evaluate the binding
ple signals present in the region of δ 0.78–4.69 also represents energies, kinetic properties and enzymes interaction for selected
straight chain structure of alkyl amines (see sup Figs. S3, S4). Spec- compounds [14]. The three dimensional coordinates of 6ND9, 2KSA
troscopic analysis of ESI-MS was used to recognize the formation and 3GHW were retrieved from RCSB Protein Data Bank (PDB);
of PGB derivative and its metal complexes. The HRMS (ESI+) cal- their respective PDB ID’s along with their affinity with α 2-δ−1,
culated for C9 H19 NO2 [M+H+] is 173.292 Da, found = 174.148 Da NK1R and DHFR receptors, respectively were tabulated in Table 1.
where as; HRMS (ESI+) calculated for compound C9H19NO2 is The molecular properties of respective ligands were deduced from
[M+H+] = 408.204 Da, and found = 408.2458 Da. This data fur- Auto Dock Tools [15–17] also tabulated in Table 1. Structure-based
ther supported the proposed structure and purity of the com- sequence configuration of α 2-δ −1, NK1R and DHFR against the se-
pounds was confirmed (sup Fig. S5). The absorption bands at 2953, lected model was performed. The 3D homology model of these re-
2598, 1641, 1386, 1332 and 1162 cm−1 confirmed the comport- ceptors in homo-sapiens was predicted using Auto Dock Tools1.5.6.
ment of -CH, -OH, -NH2, -CH3,=CH2 and C-O, respectively. How- High quality models demands an accurate sequence alignment be-
ever, 1HNMR data of compound 3 showed a doublet at δ = 0.78 tween the model and the template protein. Auto Dock generates
for CH3 functional group and also showed a clear singlet at δ = 10 clusters, the cluster with least binding energy was found to

2
A. Asbat, F. Saleem, S. Najm et al. Journal of Molecular Structure 1264 (2022) 133234

Table 1
Binding properties of receptor proteins to different ligands.

Molecular Active Receptors Interacting Efficacy Least Binding Inhibition Torsional No. of Interacting residues
Formula compound Protein(PDB energy (kcal/mol) Constant energy hydrogen
ID) (μM) bonds

C9H19NO2 3 α 2-δ −1 6ND9 Anticonvulsant −5.41 125.7 0.89 1 LEU72, ARG73, CYS81,
GLN102, MET103,
CYS104, GLN105,
TYR118,
C8H17NO2 PGB α 2-δ −1 6ND9 Anticonvulsant −5.19 157.4 0.6 1 ARG73, MET103,
CYS104, GLN105,
TYR118
C9H19NO2 3 NK1R 2KSA Anti-Inflammatory −6.41 20.15 0.89 1 LEU5, PRO6, VAL7,
THR173, MET174,
TRP184, PRO185
C13H18O2 IBU NK1R 2KSA Anti-Inflammatory −6.47 31.63 1.19 1 VAL66, THR65,
ARG130, ALA133,
HIS136, LEU138,
ARG141, LEU142,
LYS243
C9H19NO2 3 DHFR 3 ghw Anti-cancer −6.73 62.93 0.89 0 ASP21, LEU22, PHE34,
THR56, SER59, ILE60,
VAL115
C27H29NO11 DOX DHFR 3 ghw Anti-cancer −6.96 7.94 0.89 0 ILE7, VAL8, ILE16,
GLY17, ASP21, LEU22,
GLU30, PHE31, PHE34,
SER59, PRO61, ASN64,
GLY116, GLY117,
TYR121

Fig. 1. (A) Docked structure of PGB shown in orange color; with modelled 6ND9
receptor with interacting residues shown in blue color; blue dashes show hydrogen
bonding calculated by PLP. (B) The 2D interacting diagram of ligand prepared from Fig. 2. (A) Docked ligand (3) was shown in orange color with blue colored amino
protein plus online server. (C) Ligand-receptor interaction calculated by auto dock; acid residues (GLN105, and TYR118) in the binding pocket of 6ND9 calculated by
the binding energy of docked complex was found to be −5.19 Kcal/mole implies PLP, the binding energy of docked complex was found to be −5.41 Kcal/mol implies
more stable structure. more stable structure. (B) Two dimensional binding interaction of ligand show one
hydrogen bond at receptor site. (C) Docked structure of 3 with modelled 6ND9 re-
ceptor for anticonvulsant activity with interacting residues calculated by Auto Dock.

be more stable for pharmacological assessment. Structural assess-


ments and authentication of protein model were performed by the
programs called Pyrex, Protein Plus and Protein Ligand Interaction
Profiler (PLP) online servers [18–20] binding site prediction tools.
The main interacting residues at 6ND9 receptor protein with stan-
dard antiepileptic drug PGB are ARG73, MET103, CYS104, GLN105,
and TYR118 as shown in Fig. 1; with the best dock binding en-
ergy of −5.19 kcal/mol as cleared from Table 1. Whereas; the main
interacting residues of compound 3 at 6ND9 receptor are LEU72,
ARG73, GLN102, MET103, CYS104, GLN105, TYR118, and CYS81 as
revealed in Fig. 2. Receptor protein binding information of com-
pound 3 is provided in Table 1.
The main interacting residues of 2KSA protein with stan- Fig. 3. (A) Docked structure of standard IBU with modelled NK1R receptor with
interacting residues by Auto Dock for anti-inflammatory activity. (B) The 2D inter-
dard anti-inflammatory drug IBU are VAL66, THR65, ARG130,
acting diagram of ligand prepared from protein plus online server at 2KSA receptor
ALA133, HIS136, LEU138, ARG141, LEU142, and LYS243 as shown protein.
in Fig. 3. The best docked conformation of IBU has binding en-

3
A. Asbat, F. Saleem, S. Najm et al. Journal of Molecular Structure 1264 (2022) 133234

Fig. 4. (A) Compound 3 shown in orange color with interacting residues of NK1R shown in blue color in the binding pocket of 2KSA receptor calculated by online PLP
web server. (B) 2D interacting diagram of 3 showing hydrogen bond in the binding pocket of 2KSA enzyme. (C) Docked structure of 3 with modelled 2KSA enzyme with
interacting residues by Auto Dock for anti-inflammatory activity.

Fig. 5. (A) DOX was shown in orange color with blue colored amino acid residues in the binding pocket of 3GHW; calculated by PLP online server. (B) Docked structure of
DOX with modelled 3GHW receptor with interacting residues calculated by Auto Dock for anticancer potential.

ergy of −6.47 kcal/mol with 1 hydrogen bond. The main interacting


residue of potent PGB compound (3) with 2KSA receptor are LEU5,
PRO6, VAL7, THR173, MET174, TRP184, and PRO185 as shown in
Fig. 4. Binding information of 3 and IBU is provided in Table 1. The
best dock conformation of this ligand has −6.41 Kcal/mol bind-
ing energy with 1 hydrogen bond. The main interacting residues of
3GHW protein with standard anticancer drug DOX are ILE7, VAL8,
ILE16, GLY17, ASP21, LEU22, GLU30, PHE31, PHE34, SER59, PRO61,
ASN64, GLY116, GLY117, and TYR121 as shown in Fig. 5. The best
docked conformation of DOX has binding energy of −6.96 kcal/mol
with 1 hydrogen bond. The main interacting residue of potent PGB
ester (3) with 3GHW receptor are ASP21, LEU22, PHE34, THR56, Fig. 6. (A) Potent ligand 3 was shown in orange color with blue colored amino
SER59, ILE60, and VAL115 as revealed in Fig. 6. The best dock con- acid residues in the binding pocket of 3GHW; blue line show hydrogen bonding
formation of this ligand has −6.73 Kcal/mol binding energy with whereas gray dotted line represents hydrophobic interactions; calculated by PLP on-
no hydrogen bond. Receptor binding information about DOX and 3 line server. (B) Docked structure of 3 with modelled 3GHW receptor with interact-
ing residues in the binding pocket of DHFR calculated by Auto Dock.
is tabulated in Table 1.

3. Biological screening hibited substantial anti-inflammatory potential, with Inhibi-


tion/Stimulation 84.2% and IC50 = 7.7 μM. On the other hand,
3.1. Anti-inflammatory activity other compounds displayed no activity. As it is cleared from Table
S3 and Fig. 7; when synthesized metallic compounds were com-
Overall many complexes show varied anti-inflammatory activ- pared with standard ibuprofen; copper complex of PGB ester (4b)
ity. Among them, silver complexes depict better anti-inflammatory showed excellent activity, whereas, compounds 3 and 4a were in-
effect [9]. In the current research, the compound (4b) ex- active against oxidative burst assay.

4
A. Asbat, F. Saleem, S. Najm et al. Journal of Molecular Structure 1264 (2022) 133234

the study climaxes the metal co-ordination with pregabalin for


high efficacy and increased therapeutic potential. Resultantly, these
compounds will prove themselves more fruitful in future research
era in terms of their therapeutic fitness and less adverse effects.

6. Experimental

6.1. Material and method

Pregabalin (PGB) was gifted by Briell Pharmaceuticals Pvt. Ltd.,


Lahore- Pakistan. The chemicals and solvents used in research were
purchased from Sigma Aldrich (Germany). IR-spectra of conjugate-
metal complexes were recorded by Agilet FTIR spectrophotome-
ter. 1 H Nuclear magnetic resonance (NMR) spectra were recorded
on a Bruker Avance-300 spectrometer (300 MHz) by dissolving in
Fig. 7. Anti-inflammatory potential of PGB derivative and their metal complexes dimethyl sulfoxide (DMSO). Electrospray Ionization Mass spectra
shown with positive values show significant activity. (ESI-HR-MS, +/-) were measured at Bruker Compass Data Analyzer
4.2. Three dimensional crystal structures of enzymes were obtained
from Protein Data Bank. Likewise, 3D of ligand and metal com-
plexes were drawn and optimized by ACD/Chem Sketch software.
Molecular docking by Auto Dock Tools 1.5.6 and biological activi-
ties were also performed on analogues.

6.2. Synthesis

6.2.1. General procedure for synthesis of compound 3


Pregabalin (2 M) was mixed with formaldehyde (2 M) in 20 ml
ethanol. Few drops of glacial acetic acid were added as catalyst.
The mixture was stirred with magnetic stirrer for 7 to 8 h. The
progress of reaction was monitored by TLC and the final product
was evaporated at room temperature in petri dish placed in desic-
cator [12].

6.2.2. Methyl 3-(amino methyl)−5-methylhexanoate (3)


Yield: 70%; M. P.: 130 °C; IR ʋ (cm−1): 2953(-CH), 2598 (-OH),
1641, 1543 (-NH2), 1386 (-CH3), 1332 (-CH2), 1162, 1101 (C-0). 1H
Fig. 8. Cytotoxic activity (HeLa cell line) shown by different groups.
NMR (300 MHz, D2O) δ (ppm): δ 0.78 (d, J = 6.60 Hz, 3H, CH3),
1.10 (dt, J = 7.69, 7.20 Hz, 1H, CH), 2.24 (d, J = 6.94 Hz, 1H, CH),
2.86 (d, J = 7.40 Hz, 1H, CH), 4.69 (3H, s, NH2); HRMS (ESI+) cal-
culated for C9H19NO2 [M + H+] = 173.292 was found 174.148.
3.2. Anticancer (Cytotoxic) activity
6.2.3. General procedure for synthesis of metal complexes of
Different derivatives and metal complexes of pregabalin pos-
pregabalin derivatives (4a, b)
sess anti-cancer activity with their respected growth prolifera-
20 ml of methanolic solution of pregabalin derivative having
tion rates. As it is cleared from data in the Table S4 and Fig. 8
concentration of 2 mM was mixed with same quantity of methano-
that when we compared standard doxorubicin with synthesized
lic solution of ZnCl2 (2 mM) in a round bottom flask with con-
metal complexes such as PGB-LF-Cu ester (4b) showed some In-
stant stirring. The precipitates were then filtered off, washed with
hibition/Stimulation = 35.9% whereas; other compounds appeared
methanol and dried in desiccator. Same procedure was repeated
to be inactive against Hela cell lines.
with Cu(CH3COO)2. (2 mM) instead of ZnCl2. Moreover, ester Cu
complex was refrigerated for overnight [20,21].
4. Statistical analysis
6.2.4. Methyl 3-(amino methyl)−5-methylhexanoate zinc chloride
Data was expressed as mean standard deviation (±S.E), where (4a)
appropriate one-way ANOVA followed by post-hoc Dunnet’s test Yield: 70%; M. P.: 220 °C. 1H NMR (300 MHz, CDCl3) δ (ppm):
was employed. Values P < 0.5 were considered as significant. δ 3.55 (3H, s, CH3), 4.70 (3H, s, NH). 1HNMR data of compound
showed singlet peak at δ = 3.55 for CH3 functional group and also
5. Conclusion showed a clear singlet at δ = 4.70 which indicated the presence of
NH group and hence this spectroscopic analysis shows the confir-
We have synthesized esters of pregabalin and their complexes mation of FZC/4a.
with heavy metals like Zn and Cu. Few of them possessed signif-
icant anti-inflammatory potential. Furthermore, all controlled pa- 6.2.5. Methyl 3-(amino methyl)−5-methylhexanoate copper acetate
rameters and their efficacies were validated via their characteriza- (4b)
tion by different spectroscopic analysis techniques. Yield: 90%; M. P.: 230 °C; IR ʋ (cm−1 ): 3266.8 (NH), 2963.9,
Computational studies have also shown a good correlation be- 2896.1 (-CH), 1669.8 (C=O), 1546.8 (-NH2), 1485, 1459 (-CH3),
tween experiments and estimated binding energy. Considering the 1388.4,1332 (-CH2), 1276.6 (C-O). HRMS (ESI+) calculated for
increased interest towards the utilization of these metal complexes, C18H36CuN2O4 [M+H+] = 408.204, found = 408.245.

5
A. Asbat, F. Saleem, S. Najm et al. Journal of Molecular Structure 1264 (2022) 133234

6.3. Solubility studies this, HeLa cell line was cultured in Minimum Essential Medium Ea-
gle, accompanied with 5% of fetal bovine serum (FBS), 100 IU/ml
Apparent solubility of all the synthesized metals was deter- of penicillin and 100 μg/ml of streptomycin in 75 cm2 flasks, and
mined. A measured amount of synthesized compounds were dis- preserved in 5% CO2 incubate at 37 °C. Additionally, exponen-
solved in weighed amount of solvent and clarity of solution was tially growing cells were harvested, calculated with hemocytome-
observed. Water, methanol, ethanol, DMSO and chloroform were ter and diluted with a respective media. Cell culture possessing
used for this study. For good solubility the threshold value of the concentration of 6 × 104 cells/ml was prepared and trans-
<0.1/100 ml with the clear colored solution in the given solvent ferred (100 μl/well) into 96-well plates. Medium was removed af-
was considered [13]. ter overnight incubation, and 200 μl of fresh medium was added
with varied concentrations of compounds (1–30 μM). However,
6.3.1. Molecular docking studies 200 μl MTT (0.5 mg/ml) was added in all wells individually after
Docking model was based on 3D crystal structure of α 2-δ −1 48 h and then incubated additionally for 4 h. Successively, 100 μl
subunit (PDB-ID: 6ND9) (https://www.rcsb.org/ structure/6ND9) of DMSO was added to each well separately. The degree of MTT re-
for anti-convulsant activity, neurokinin-1 receptor (PDB-ID: 2KSA) duction to formazan within cells was examined by determining the
(https://www.rcsb.org/structure/2KSA) for anti-inflammatory activ- absorbance at 570 nm, using a micro plate reader. Finally, the cy-
ity and dihydrofolate reductase receptor (PDB-ID: 3GHW) (https:// totoxicity was calculated as concentration causing 50% growth in-
www.rcsb.org/structure/3GHW) for antitumor activity; were re- hibition (IC50) for HeLa cell line [25,26].
trieved from RCSB Protein Data Bank (PDB). ACD/Chemsketch soft- The percent inhibition was calculated by following formula:
ware was used for the 3D structure buildup of various ligands and % age inhibition=100-((mean of O.D of test compound-mean
save as Mol file; Open Babel-GUI 3.0 program was used to convert
of O.D of negative control)/(mean of O.D of positive control-
Mol file to PDB file for automated docking by Auto Dock Tools 1.5.6
software [16]. The docking protocol was tested by removing the mean of O.D of negative control) *100).
co-crystallized inhibitor from the protein and docks it on the same Doxorubicin% inhibition at 30μ = 101.2%
side. Protein structure was added by polar hydrogens and gestesian Doxorubicin IC50 = 0.9 ± 0.14
charges during protein preparation by Auto dock Tools graphical
user interface. The cuboid grid box was used for the computation
with a purpose to embrace all the minimized inhibitors spanning Declaration of Competing Interest
10 A˚ in all three dimensions. The Lamarckian genetic algorithm
(LGA) was used to generate conformations of ligands within the The authors declare that they have no known competing finan-
binding site, with an initial population size of 150 individuals, with cial interests or personal relationships that could have appeared to
a maximum number of 250,0 0 0 energy evaluations, 150,0 0 0 gen- influence the work reported in this paper.
erations with a mutation rate of 0.02, and a cross-over rate of 2
CRediT authorship contribution statement
points. The rigidity parameters were set for the receptor, keeping
the ligand flexible. Out of the ten docked conformations obtained,
Ayesha Asbat: Data curation, Formal analysis, Investigation,
one having the lowest binding energy was selected andthe active
Methodology, Software, Writing – original draft, Funding acqui-
site of enzyme. Root mean square deviation (RMSD) was calculated
sition, Writing – review & editing. Farooq Saleem: Data cura-
and in all cases RMSD value of <2.0 Å was considered significant
tion, Formal analysis, Methodology, Supervision, Validation, Writ-
in predicting binding orientation of ligand [11].
ing – review & editing. Saima Najm: Conceptualization, Data
curation, Formal analysis, Investigation, Methodology, Resources,
6.4. Biological screenings Software, Supervision, Validation, Visualization, Writing – original
draft, Writing – review & editing. Javed Iqbal: Data curation, Fund-
6.4.1. Anti-inflammatory activity ing acquisition, Writing – review & editing. Muhammad Ali Syed:
In-vitro anti-inflammatory activity of ligand esters and their Data curation, Supervision, Writing – review & editing. Sarfraz Ah-
metal complexes were evaluated (7–9). Luminol-enhanced Chemi- mad: Writing – review & editing. Sana Hanif: Formal analysis, In-
luminescence assay was performed, as described by Helfand et al. vestigation, Writing – review & editing. Muhammad Azeem: Data
[22]. For this purpose, 25 μl of diluted whole blood HBSS++ (Hanks curation, Investigation, Writing – review & editing.
Balanced Salt Solution, containing calcium chloride and magne-
sium chloride) was incubated with 25 μl of three varied concen- Acknowledgement
trations of compounds (1, 10 and 100 μg/ml), each in triplicate.
Both (HBSS++ and cells having no compounds) were introduced The authors are grateful Muhammad Mubbashir Khan (Quality
into control wells. Test was implemented in white half area of 96 Control Manager), Briell Pharmaceuticals (Pvt.) Ltd, Lahore-Pakistan
well plates and was incubated at 37 °C for 15 min in the ther- for his skillful assistance in recording FTIR. Thanks are also due
mostat chamber of luminometer. Subsequently, 25 μl of serum op- to Quaid-e-Azam University Islamabad for characterization of com-
sonized zymosan (SOZ) and 25 μl of intracellular reactive oxygen pounds and International center for Chemical and Biological sci-
species detecting probe, luminol were added into all wells individ- ences (Karachi) for ESI-MS and in-vitro (anti-inflammatory and
ually except the blank which possessed only HBSS++. The level of anti-cancer) studies of synthesized chemical compounds.
the reactive oxygen species (ROS) was examined in luminometer in
the form of relative light units (RLU) (10, 11). Moreover, standard Supplementary materials
drug used for the assay was Ibuprofen with IC50 = 11.2 ± 1.9 μM
[22–24]. Supplementary material associated with this article can be
found, in the online version, at doi:10.1016/j.molstruc.2022.133234.
6.4.2. Anticancer activity
References
Standard MTT (3-[4, 5-dimethylthiazole-2-y1]−2, 5-diphenyl-
tetrazolium bromide) 96 well plates colorimetric assay was used [1] B.A. Leeman, A.J. Cole, Advancements in the treatment of epilepsy, Annu. Rev.
to assess the cytotoxic activity of pregabalin derivatives. To achieve Med. 59 (2008) 503–523, doi:10.1146/annurev.med.58.071105.110848.

6
A. Asbat, F. Saleem, S. Najm et al. Journal of Molecular Structure 1264 (2022) 133234

[2] A. Bali, P. Gaur, A novel method for spectrophotometric determination of pre- [15] F. Abid, S. Najm, In silico appraisal of lupeol and butelinic acid as compelling
gabalin in pure form and in capsules, Chem. Cent. J. 1 (2011) 1–7, doi:10.1186/ anti inflammatory mediators, Int. J. Biol. Pharm. Allied. Sci 8 (2019) 1433–
1752- 153X- 5- 59. 1443.
[3] S. Hu, C.A. Martinez, J. Tao, Preparation of pregabalin and related compounds, [16] S. Shukla, A.P. Mishra, Synthesis, structure, and anticancerous properties of sil-
Epilepsy Res. 1 (2012) 1–38. ver complexes, J. Chem. 13 (2013) 1–6, doi:10.1155/2013/527123.
[4] Z. Haneef, H.S. Levin, S. Chiang, Brain graph topology changes associated with [17] W.M.E. Kayal, S. YuShtrygo, S.V. Zalevskyi, A. Shark, V.V. Tsyvunin, S.M. Ko-
anti-epileptic drug use, Brain Connect 1 (2015) 1–43, doi:10.1089/brain.2014. valenko, N.D. Bunyatyan, L.O. Perekhoda, H.I. Severina, V.A. Georgiyants, Syn-
0304. thesis, in vivo and in silico anticonvulsant activity studies of new derivatives
[5] V. Verma, N. Singh, A.S. Jaggi, Pregabalin in neuropathic pain: evidences and of 2-(2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl) acetamide, Eur. J. Med. Chem.
possible mechanisms, Curr. Neuropharmacol. 12 (2014) 44–56, doi:10.2174/ 180 (2019) 134–142, doi:10.1016/j.ejmech.2019.06.085.
1570159X1201140117162802. [18] G.M. Morris, R. Huey, W. Lindstrom, M.F. Sanner, R.K. Belew, D.S. Good-
[6] J.B. Schwarz, S.E. Gibbons, S.R. Graham, N.L. Colbry, P.R. Guzzo, M.G. Vartanian, sell, J. Olson, AutoDock4 and autoDockTools4: automated docking with selec-
J.J. Kinsora, S.M. Lotarski, Z. Li, M.R. Dickerson, T. Su, M.L. Weber, A.J. Thorpe, tive receptor flexibility, J. Comp. Chem. 30 (2009) 2785–2791, doi:10.1002/jcc.
S.D. Donevan, C.P. Taylor, D.J. Wustrow, Novel cyclopropyl δ -amino acid ana- 21256.
logues of pregabalin and gabapentin that target the r2-δ protein, J. Med. Chem. [19] A. Asbat, S. Najam, J. Iqbal, M. Mubbashir, M. Ali, S. Khurshid, Metal
48 (2004) 3026–3035, doi:10.1021/jm0491086. complexes of Pregabalin: design, synthesis, characterization and biological
[7] M. Kapucu, Enantioselective formal synthesis of pregabalin with bifunctional evaluation, J. Mol. Struct. 1235 (2021) 130236, doi:10.1016/j.molstruc.2021.
acid/base organocatalysts, Eur. J. Org. Chem. 1 (2013) 1–78. 130236.
[8] B. Frame, R. Miller, M.M. Hutmacher, Joint modeling of dizziness, drowsiness, [20] S. Zazouli, L. Laallam, E. Mostafa, Synthesis of novel benzohydrazide and ben-
and dropout associated with pregabalin and placebo treatment of general- zoic acid derivative: crystal structure, hirshfeld surface analysis and DFT com-
ized anxiety disorder, J. Pharmacokinet. Phar. 36 (2009) 565–584, doi:10.1007/ putational studies, J. Mol. Struct. 1239 (2021) 130465, doi:10.1016/j.molstruc.
s10928- 009- 9137-5. 2021.130465.
[9] L. Kyros, N. Kourkoumelis, M. Kubicki, L. Male, M.B. Hursthouse, I. I.Verginadis, [21] K. Salat, T. Librowski, B. Nawiesniak, L.M. Gluch, Evaluation of analgesic, an-
E. Gouma, S. Karkabounas, K. Charalabopoulos, S.K. Hadjikakou, Structural tioxidant, cytotoxic and metabolic effects of pregabalin for the use in neuro-
properties, cytotoxicity, and anti-inflammatory activity of silver(I) complexes pathic pain, Neurol. Res. Paper 35 (2013) 948–958, doi:10.1179/1743132813Y.
with tris(p-tolyl) phosphine and 5-Chloro-2-mercaptobenzothiazole, Bioinorg. 0 0 0 0 0 0 0236.
Chem. Appl. (2010) 1–12, doi:10.1155/2010/386860. [22] S.L. Helfand, J. Werkmeister, J.C. Roder, Chemiluminescence response of human
[10] D.C. Reis, M.C.X. Pinto, E.M.S. Fagundes, L.F. Rocha, V.R.A. Pereira, C.M.L. Melo, natural killer cells, J. Exp. Biol. 156 (1982) 492–504, doi:10.1084/jem.156.2.
H. Beraldo, in: Investigation on the Pharmacological Profile of Antimony (III) 492.
Complexes With Hydroxyquinoline derivatives: Anti-Trypanosomal Activity and [23] A.O. Oladimeji, I.A. Oladosu, A. Jabeen, A. Faheem, M.A. Mesaik, M.S. Ali, Im-
Cytotoxicity Against Human Leukemia Cell Lines, Springer Science+Business munomodulatory activities of isolated compounds from the root-bark of Cus-
Media, 2011, pp. 9407–9408, doi:10.1007/s10534- 011- 9407- 8. sonia arborea, Pharm. Biol. 55 (2017) 2240–2247, doi:10.1080/13880209.2017.
[11] A.S. Thakur, R. Deshmukh, A.K. Kumar, Molecular docking study and 140 0 078.
anticonvulsant activity of synthesized 4-((4,6-dimethyl-6H-1,3-thiazin-2- [24] D. Demertzi, D. Hadjipavlou-Litina, M. Staninska, A. Primikiri, C. Kotoglou,
yl)phenylsulfonyl)urea/thiourea derivatives, J. King. Saud. Univ. Sci. 30 (2018) M.A. Demertzis, Anti-oxidant, in vitro, in vivo anti-inflammatory activity and
330–336, doi:10.1016/j.jksus.2016.12.006. antiproliferative activity of mefenamic acid and its metal complexes with man-
[12] A. Xavier, N. Srividhya, Synthesis and study of Schiff base ligands, J. Appl. ganese(II), cobalt(II), nickel(II), copper(II) and zinc(II), J. Enzyme Inhib.Med.
Chem. 7 (2014) 06–15, doi:10.9790/5736-071110615. Chem. 24 (2009) 742–752, doi:10.1080/14756360802361589.
[13] M. Jabeen, S. Ahmad, K. Shahid, A. Sadiq, U. Rashid, Ursolic acid hydrazide [25] T. Mosmann, Rapid colorimetric assay for cellular growth and survival: appli-
based organometallic complexes: synthesis, characterization, antibacterial, an- cation to proliferation and cytotoxicity assays, J. Immunol. Methods (1983) 55–
tioxidant, and docking studies, J. Front. Chem. 6 (2018) 55–65, doi:10.3389/ 63, doi:10.1016/0022- 1759(83)90303- 4.
fchem.2018.0 0 055. [26] K.A. Ali, M.M. Abd-Elzaher, K. Mahmoud, Synthesis and anticancer properties
[14] N. Shahabadi, S. Amiri, A. Taherpour, Human serum albumin binding studies of silver(I) complexes containing 2,6-Bis(substituted)pyridine derivatives, Int. J.
of a new platinum(IV) complex containing the drug pregabalin: experimental Med. Chem. (2013) 256836.
and computational methods, J. Coord. Chem. 72 (2019) 600–618, doi:10.1080/
00958972.2019.1568419.

You might also like