You are on page 1of 11

Journal of Molecular Structure 1255 (2022) 132482

Contents lists available at ScienceDirect

Journal of Molecular Structure


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

Synthesis, in vitro anticancer activity and reactions with biomolecule


of gold(I)-NHC carbene complexes
Sughra Gulzar a, Umme Ammara a, Zeeshan Abid a, Munazza Shahid b, Raja Shahid Ashraf a,
Nadeem Baig c, Abdel-Nasser Kawde d, Gaurav Bhatia e, Anvarhusein A. Isab f,∗,
Muhammad Altaf a,∗
a
Department of Chemistry, Government College University Lahore, Pakistan
b
Department of Chemistry, University of Education, Lahore, Pakistan
c
Interdisciplinary Research Center for Membranes and Water Security, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
d
Department of Chemistry, College of Sciences, Research Institute of Sciences and Engineering, University of Sharjah, P.O. Box 27272, Sharjah, United Arab
Emirates
e
Department of Biochemistry, Pandit Jawarharlal Medical College and Hospital, Chamba, Himachal Pradesh 176310, India
f
Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia

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

Article history: Gold(I) N-heterocyclic carbene (NHC) complexes are widely investigated for promising anticancer activ-
Received 20 October 2021 ities. Herein, we present the synthesis, structural elucidation and in vitro cytotoxic profiles of five new
Revised 20 January 2022
gold(I)-NHC complexes containing thione ligands represented as [Au(IPr)(thione)]PF6 (where IPr = 1,3-
Accepted 22 January 2022
Bis(2,6-diisopropylphenyl)imidazol-2-ylidene and thione refers to 1,3-imidazolidine-2-thione (Ims) and its
Available online 24 January 2022
four N-mono- and N,N’-dialkyl (alkyl = -CH3 , -C2 H5 ) derivatives. Structural confirmation of all synthesized
Keywords: compounds has been carried out by elemental analysis, FTIR, 1 H NMR and 13 C NMR spectroscopy. Three
Gold out of five complexes (1, 3, 4) have also been subjected to single-crystal X-ray diffraction studies, which
N-heterocyclic carbene revealed the presence of [Au(IPr)(thione)]+ and PF6 − as counter ions and close to linear coordination
Thione geometry of gold(I) atoms. In vitro antitumor studies of synthesized complexes were examined against
X-ray diffraction analysis human cancer cell lines of colon, lung and breast cancers, namely HCT-15, A549 and MCF7 using MTT
In vitro anticancer activity
assay. The electrochemical reactions were also performed to know the possible mechanism of interaction
Biomolecule
between complexes and biomolecule.
© 2022 Elsevier B.V. All rights reserved.

1. Introduction platinum-containing antitumor medicines like carboplatin and


oxaliplatin share the same complications [6]. These limitations
Transition metal-based complexes have earned a repute as tempted researchers to investigate non-platinum metal complexes
potential therapeutics against ancient and contemporary ailments that offer good antitumor properties while being safer for cancer
caused by pathogens and biological abnormalities [1–3]. A prime subjects. Therefore, a large number of studies have undertaken
example of the prevailing medicinal benefits of transition metal gold, silver, ruthenium, rhodium, iron, copper, cobalt, palladium
complexes is Cisplatin, a platinum-based complex that currently and some other metals as an alternative to platinum metal [7–15].
stands as the most valued anticancer drugs for chemotherapy Consequently, several metal complexes comprising palladium,
against many types of cancers [4]. Although cisplatin is licensed ruthenium and gold metals have been approved as antibacte-
for medical use since the 1970s and appears in the WHO’s List rial, antifungal, antiinflammatory, antirheumatic and antitumour
of Essential Medicines, it has a number of side effects including agents [16].
nephro-, neuro- and ototoxicity [5]. Coupled with the post- The healing power of gold has been valued since early human
treatment recurrence of the cisplatin-resistant disease in patients, history. In conventional Egyptian and Asian medicines, gold com-
these side effects hinder the efficiency of cisplatin. Similarly, other plexes have been utilized for treating tuberculosis, inflammation
and infection [17]. In modern history, the initial healing studies
of gold complexes were focused on inflammatory ailments. In the

Corresponding authors.
early 1920s, gold(I)-thiolate and gold(I)-phosphine complex (Aura-
E-mail addresses: aisab@kfupm.edu.sa (A.A. Isab), muhammad.altaf@gcu.edu.pk nofin) were utilized to cure rheumatoid arthritis [18,19]. With ad-
(M. Altaf). vances in the research, it was observed that these gold complexes

https://doi.org/10.1016/j.molstruc.2022.132482
0022-2860/© 2022 Elsevier B.V. All rights reserved.
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

show low indications of malignancy rates. This observation raised 2. Experimental


the assumption that gold complexes might possess antitumour ac-
tivity. In the early 1970s and 1980s, numerous groups of gold(III) 2.1. Materials
compounds emerged as promising cytotoxic agents [20]. Many gold
complexes evaluated for antitumor activity comparable with plat- Common chemicals i.e., ethyl alcohol (C2 H5 OH) and dichloro-
inum(II) complex (cisplatin) have shown better potency for can- methane (CH2 Cl2 ) were bought from local commercial sources.
cer treatment through different mechanisms, including inhibition 1,3-bis(2,6-diisopropylphenyl)imidazol-2-ylidenegold(I)chloride)
of thioredoxin reductase or proteasome, direct DNA damage and (Au(IPr)Cl), silver hexafluorophosphate (AgPF6 ) and L-tyrosine
alteration of cell cycles [21–24]. were procured from Sigma-Aldrich, St. Louis, United States. Dul-
In addition, researchers have emphasized the significant role of becco’s Modified Eagle Medium (DMEM) and HCT-15, A549 and
the ligands especially because of their ability to tune cytotoxicity, MCF7 cell lines were obtained from The National centre for Cell
lipophilicity, stability and specificity of the complexes [25,26]. The Science, Pune, India. 2H-Imidazole-2-thione (ImS) and its four dif-
choice and role of ligands are of crucial importance for the effec- ferent alkyl derivatives were synthesized and purified by literature
tiveness, selectivity and potency of gold complexes. Several gold(I) procedures [67,68]. Double-distilled water (ddH2 O) was acquired
and gold(III) complexes containing thiolate ligands have exhibited from Aquatron A40 0 0D WaterStill assembly.
higher anticancer potential than cisplatin [27–32]. Among these,
gold(I)-phosphine thiolate complexes are particularly significant 2.2. Instrumentation
owing to a higher cytotoxic activity than simple gold(I)-thiolates.
Anticancer efficiency of gold(I)-phosphino-dithiocarbamate com- Elemental analyses data was obtained from Perkin-Elmer 2400
plexes proved that P−Au−S moiety contributes to a higher cyto- Series II (CHNS/O), Analyzer. Solid-state FTIR analysis was con-
toxic potential of complexes. It has also been proven that phos- ducted on Perkin-Elmer FTIR 180 spectrophotometer using KBR
phine ligands enhanced the membrane permeability and lipophilic- pellets of samples over the range 40 0 0–40 0 cm−1 and 4 cm−1 res-
ity of the gold(I)-phosphine complexes that contribute to their an- olution. LAMBDA NMR spectrophotometer (500 MHz) was used for
ticancer character [33,34,35]. The studies have shown that gold(I)- the recording of 13 C (125.65 MHz),1 H (500.01 MHz) NMR analy-
phosphine imidazolates [36] and pyrazolates complexes have good sis. Tetramethylsilane was used as internal standard and chemical
solubility and are seventy times more active against lung, human shifts were presented in δ (ppm) relative to it. NMR spectra were
breast, cervical colon and ovarian cancer cell lines than cisplatin obtained at 297 K using chloroform-d (CDCl3 ) as solvent. The X-ray
[37–39]. The studies have also proved that the right choice of lig- data of all complexes were taken from STOE’s IPDS II containing a
and can take the therapeutic potential of gold(I) complexes beyond two-circle Goniometer and graphite-monochromated MoKα radia-
cancer treatment. Additionally, proven activity of gold(I) complexes tion (λ = 0.71073 Å) at 173 K (-100 °C). SHELXS-2014 program was
against rheumatoid arthritis [40], gold benzimidazole derivatives employed to resolve and refine the structures using direct meth-
have shown antileishmanial and antibacterial activity [41,42]. Like- ods. A semi-empirical method implemented by MULABS in PLA-
wise, gold complexes are anticipated to bear therapeutic activity TON was used for absorption correction. Cyclic Voltammetry (CV)
against HIV and AIDS [43,44]. and Square Wave Voltammetry (SWV) calculations were produced
Over the last few years, extensive research has been carried out using AutoLab (Netherlands). The electrochemical workstation con-
on imidazole- and benzimidazole like structures because of their sisted of working glassy carbon electrode, platinum counter elec-
presence in many potent therapeutic agents [45,46]. Some of the trode and silver chloride (Ag-AgCl) reference electrode. Accumet®
common anticancer drugs containing imidazole moieties include XL50 meter was utilized to monitor buffer pH. GR-20 0 0 electrical
dacarbazine, zoledronate (Zometa), azathioprine (Imuran) and tipi- balance was used for the measurements of the weights of the var-
farnib (Zarnestra), all contain imidazole moieties [47–49]. Similarly, ious chemicals.
many antihistaminic (cimetidine, imetit and immepip), antihy-
pertensive (eprosartan, losartan, olmesartan), antifungal (micona- 2.3. Synthesis of complexes
zole, oxiconazole, clotrimazole), antiparasitic (benznidazole, sec-
nidazole, metronidazole) compounds comprising imidazole moi- 2.3.1. Synthesis of [Au(IPr)(ImS)]PF6 (1)
eties are commonly used against different diseases with great ther- 0.500 mmol (0.311 g) of Au(IPr)(Cl) was first solubilized in
apeutic potency [46,50,51]. 15.0 mL of dichloromethane (DCM) and then added to 5.0 mL
Recently, NHCs have (Fig. 1) grabbed the attention of re- ethanolic solution of 0.500 mmol (0.127 g) of AgPF6 . The mix-
searchers as emerging ligands of imidazole and benzimidazole ture was magnetically stirred for 5 min at room temperature fol-
classes to form promising bioactive inorganic complexes [52–55]. lowed by filtration. After filtration, 0.500 mmol (0.051 g) of 1,3-
A carbene is a two-coordinate state of carbon that contains a Imidazolidine-2-thione (ImS) was added to the filtrate. After 1 h
lone pair of electrons. NHCs attain stability by forming two ni- of additional stirring, the mixture was filtered off and placed
trogen atoms and coordinate efficaciously to many metal centers in refrigerator for crystal growth. The white crystals were col-
to form stable metal-NHC complexes [56,57]. In particular, gold- lected after three to four days. Yield 57% (0.237 g). Anal. Calc.
NHC complexes are valued for their stability, catalytic properties, for C30 H42 AuN4 S·PF6 (832.68), C, 43.22; H, 5.20; N, 6.72; S, 4.65.
controllable steric hindrance, and antitumor potential [52]. Be- Found: C, 43.38; H, 5.34; N, 6.61; S, 4.32%. 1 H NMR (CDCl3 , δ ppm):
sides, gold-NHCs have also shown antibacterial activity [58], an- 8.12 (NH, s, 2H), 7.44 (3H, d, 2H), 7.59 (4H, t, 2H), 3.38 (5H, m, 4H),
tileishmanial activity [59], thioredoxin reductase (TrxR) inhibition 1.21 (6H, d, 12H), 1.25 (7H, s, 12H), 8.08 (8H, s, 2H), 3.54 (11H, s,
[60] and antiproliferative properties [61–66]. The captivating ther- 4H). 13 C NMR (CDCl3 , δ ppm):145.4 C1, 133.6 C2, 124.9 C3, 130.7
apeutic potential of gold complexes encouraged us to synthesize C4, 28.4 C5, 23.9 C6, 23.6 C7, 124.1 C8, 173.6 C9, 179.0 C10, 44.8
and examine gold(I)-NHC complexes (1–5) bearing a soft thione C11.
co-ligand which is likely to impart additional stability to the com-
plexes. The complexes were characterized using advanced analyt- 2.3.2. Synthesis of [Au(IPr)(MeImS)]PF6 (2)
ical techniques and examined for cytotoxicity against colon (HCT- It was synthesized according to the above procedure for (1)
15), lung (A549) and breast (MCF7) cancer cell lines. This study except that 0.500 mmol (0.058 g) of N-methyl-1,3-imidazolidine-
provides first-ever insight into cytotoxic profile of gold(I)-NHCs 2-thione (MeImS) was added as ligand instead of ImS and prod-
with ligands reported herein. uct was filtered as white powder. Yield: 65% (0.265 g). Anal. Calc.

2
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

for C31 H45 AuN4 S·PF6 (847.27), C, 42.92; H, 5.35; N, 6.61; S, 4.56. Table 1
Selective IR bands (cm–1 ) of ligands and complexes.
Found: C, 42.93; H, 5.44; N 6.88; S,4.53%. 1 H NMR (CDCl3 , δ ppm):
8.10 (NH, s, 1H), 7.44 (3H, d, 2H), 7.55 (4H, t, 2H), 2.63 (5H, m, 4H), Compound ν (C=S) ν (N–H) ν (C–H)
1.23 (6H, d, 12H), 1.22 (7H, s, 12H), 8.11 (8H, s, 2H), 3.56 (11H, s, [Au(IPr)Cl] – – 3073, 2960
2H), 3.37 (12H, s, 6H). 13 C NMR (CDCl3 , δ ppm):145.3 C1, 133.6 C2, ImS 510, 1199 3200 –
124.9 C3, 130.7 C4, 28.4 C5, 23.9 C6, 23.4 C7, 130.7 C8, 173.5 C9, (1) 496, 1061 3377 3169, 2964, 2869
178.2 C10, 48.8 C11, 34.3 C12. MeImS 520, 1200 3200 –
(2) 470, 1058 3360 2962, 2873
Me2 ImS 516, 1201 – –
2.3.3. Synthesis of [Au(IPr)(Me2 ImS)]PF6 (3) (3) 496, 1123 – 2963, 2870
It was synthesized according to the above procedure for (1) ex- EtIms 515 3200 –
cept that 0.500 mmol (0.065 g) of N,N’-dimethyl-1,3-imidazolidine- (4) 441, 1060 3398 3120, 2962
Et2 ImS 514, 1199 – –
2-thione (Me2 ImS) was added as ligand instead of ImS. The prod-
(5) 449, 1118 – 3145, 2963
uct was obtained as white crystals. Yield: 63% (0.285 g). Anal. Calc.
for C32 H47 AuN4 S·PF6 ·C2 H6 O, (907.32), C, 44.98; H, 5.88; N, 6.17; S,
4.47. Found: C, 44.82; H, 5.71; N 6.18; S,4.23%. Yield = 0.285 g
(63%). 1 H NMR (CDCl3 , δ ppm): 7.44 (3H, d, 2H), 7.61 (4H, t, 2H), until it appeared purple. The optical density of 96 well-plates was
2.64 (5H, m, 4H), 1.23 (6H, d, 12H), 1.21 (7H, s, 12H), 8.12 (8H, s, found out using LabSystems Multiskan EX ELISA reader at 570 nm.
2H), 3.57 (11H, s, 2H), 3.37 (12H, s, 6H). 13 C NMR (CDCl3 , δ ppm): The outcomes of the study are shown as IC50 value of each com-
145.3 C1, 133.5 C2, 124.1 C3, 124.9 C4, 28.3 C5, 23.9 C6, 23.4 C7, plex which corresponds to micromolar concentration effective for
130.7 C8, 171.3 C9, 178.0 C10, 48.8 C11, 34.4 C12. 50% inhibition of cell growth.

2.3.4. Synthesis of [Au(IPr)(EtImS)]PF6 (4) 2.5. Electrochemical measurements


It was synthesized according to the above procedure for (1)
except that 0.500 mmol (0.079 g) of N-ethyl-1,3-imidazolidine-2- Ethanolic solutions of synthesized complexes were subjected to
thione (EtImS) was added as ligand instead of ImS. The product electrochemical analysis owing to their greater solubility in ethanol
was obtained as white crystals. Yield: 61% (0.244 g). Anal. Calc. compared with water. Before each analysis, bare GCE was cleaned
for C32 H46 AuN4 S·PF6 (860.72), C, 44.60; H, 5.50; N, 6.50; S, 4.47. with alumina slurry using a polishing clothe till the mirror-like re-
Found: C, 44.82; H, 5.47; N 6.43; S, 4.67%. 1 H NMR (CDCl3 , δ ppm): flection was achieved. The SWV and CV were range was set from
8.13 (NH, s, 1H), 7.10 (3H, d, 2H), 6.95 (4H, t, 2H), 2.00 (5H, m, 4H), 0.2 to 1.0 V for numerous analyses of the anticancer complexes and
0.75 (6H, d, 12H), 0.73 (7H, s, 12H), 7.62 (8H, s, 2H), 2.86 (11H, m, the L-tyrosine interaction.
2H), 3.69 (12H, m, 2H), 3.56 (13H, 2H), 0.30 (14H, m, 3H). 13 C NMR
(CDCl3 , δ ppm): 145.3 C1, 133.5 C2, 123.7 C3, 130.6 C4, 28.3 C5, 3. Results and discussion
23.9 C6, 23.5 C7, 124.1 C8, 182.0 C9, 178.4 C10, 41.1 C11, 48.1 C12,
41.6 C13, 11.6 C14. 3.1. Synthesis and spectroscopic characterization

2.3.5. Synthesis of [Au(IPr)(Et2 ImS)]PF6 (5) The elemental analysis and spectroscopic data confirmed the
It was synthesized according to the above procedure for (1) ex- formation of complexes. The synthetic scheme and molecular
cept that 0.500 mmol (0.079 g) of N,N’-diethyl-1,3-imidazolidine- structures of ligands are illustrated in Scheme 1.
2-thione (Et2 ImS) was used as ligand instead of ImS and product The IR frequencies absorbed by functional groups present in
was filtered as white powder. Yield = 61% (0.244). Anal. Calc. for complexes are presented in Table 1. The IR band of ν (C=S)
C34 H51 AuN4 S·PF6 (889.31), C, 45.98; H, 5.78; N, 6.30; S,4.30. Found: in thiones was noticed between 1191–1217 cm−1 in complexes,
C, 45.88; H, 6.28; N, 4.13; S, 4.51%. 1 H NMR (CDCl3 , δ ppm): 7.44 which justifies the successful complexation. The stretching band of
(3H, d, 2H); 7.73 (4H, t, 2H), 3.13 (5H, m, 4H), 1.21 (6H, d, 12H), ν (N–H) shifted to a higher frequency in complexes. Likewise, high
1.05 (7H, d,12H), 7.59 (8H, s, 2H), 3.44 (11H, d, 2H); 3.59 (12H, t, frequency shift of C-N bond indicated an increase in its π char-
2H), 0.89 (13H, dd, 2H). 13 C NMR (CDCl3 , δ ppm): 145.3 C1, 133.5 acter. Overall, the results conform with existing reports of gold(I)-
C2, 123.7 C3, 130.7 C4, 28.6 C5, 24.0 C6, 23.9 C7, 124.9 C8, 182.1 thione complexes [69–71].
1 H NMR spectral data (δ , ppm) of complexes is presented in
C9, 177.0 C10, 45.9 C11, 42.2 C12, 11.5C13.
Table 2. The chemical shifts for Au(IPr)Cl are similar to the pre-
2.4. In vitro anticancer activity evaluation vious reports [72]. The N-H signals of thiones in 1 H NMR shifted
downfield (higher ppm) compared with their chemical shifts in the
The cytotoxicity of synthesized complexes was evaluated free state. Upon coordination of thione with metal, transmission
against colon, lung and breast cancer cell lines employing MTT as- of electron density from N to S increase π character of C-N bond
say. HCT-15, A549 and MCF7 cells were seeded and maintained which in turn deshielded the thiones and increased the chemi-
in triplicate onto 96-well plates at 3 × 103 cells/well density in cal shift value (downfield). Table 3 presents the selected 13 C NMR
DMEM (100 μL) comprising 10% fetal bovine serum (FBS) and in- shifts of thiones. In complexes, C-C resonance shifted to a higher
cubated for growth in CO2 incubator for 72 h (37 °C, 5% CO2 chemical shift value of >6 ppm in comparison with Au(IPr)Cl. The
and 90% relative humidity). This was followed by addition of 100 downfield shift results from transfer of electron density towards
μL solution of cisplatin standard and synthesized complexes in the metal from C-N bond as the complexation takes place. The C=S
DMEM and further incubation for 24 h (37 °C, 5% CO2 ). The cul- chemical shift of [Au(IPr)(thiones)]PF6 complexes is observed up-
ture medium was removed followed by addition of 100 μL DMEM field compared with free thiones. This upfield shift conforms with
having 0.5 mg/ml MTT. Further incubation for 4 h (37 °C, 5% CO2 ) the literature data [73,74].
in dark resulted in the formation of purple-formazan crystals at
the base of wells. After careful removal of the medium without 3.2. Crystal structure determination
disrupting the monolayer of MTT metabolic product, DMSO (100
μL) was introduced to each well and the well-plates was shaken at The structural represention of [Au(IPr)(ImS)]PF6 (1),
150 rpm for 5 min for thorough dissolution of formazan crystals [Au(IPr)(Me2 ImS)]PF6 ·C2 H6 O (3) and [Au(IPr)(EtImS)]PF6 (4) is

3
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

Scheme 1. Synthetic scheme of gold(I)-NHC thione complexes and structures of ligands.

Table 2
1
H NMR spectral data (δ , ppm) of complexes.

Compound NH 3H 4H 5H 6H 7H 8H 11H 12H 13H 14H

ImS 7.90 – – – – – – 3.59 – – –


(1) 8.12 7.44 7.59 2.49 1.21 1.25 8.08 3.54 – – –
MeImS 7.93 – – – – – – 3.63 3.4 2.9 –
(2) 8.10 7.44 7.55 2.63 1.23 1.22 8.11 3.56 3.37 – –
Me2 ImS – – – – – – – 3.48 2.91 – –
(3) – 7.44 7.59 2.64 1.23 1.21 8.12 3.57 3.37 – –
EtImS 5.67 – – – – – – 3.70 3.58 3.67 1.20
(4) 8.13 7.10 6.95 2.00 0.75 0.73 7.62 2.86 3.69 3.56 0.71
Et2 ImS – – – – – – – 3.48 3.37 0.97 –
(5) – 7.44 7.73 2.44 1.21 1.05 7.59 3.44 3.59 0.89 –

Table 3
13
C NMR spectral data (δ , ppm) of complexes.

Compound C=S (C10) Au-C (C9) C1 C2 C3 C4 C5 C6 C7 C8 C11 C12 C13 C14

ImS 182.11 – – – – – – – – – 45.38 – – –


(1) 179.0 173.6 145.4 133.6 124.9 130.7 28.4 23.9 23.6 124.1 44.8 _ – –
MeImS 181.38 – – – – – – – – – 42.00 51.82 34.35 –
(2) 178.2 173.5 145.3 133.6 124.9 130.7 28.4 23.9 23.4 130.7 48.8 34.3 – –
Me2 ImS 180.46 – – – – – – – – – 48.77 48.77 34.91 –
(3) 178.0 171.9 145.3 133.5 124.1 124.9 28.3 23.9 23.4 130.7 48.8 34.4 – –
EtImS 182.90 – – – – – – – – – 40.68 47.03 40.41 11.80
(4) 178.4 178.4 145.3 133.5 123.7 130.6 28.3 23.9 23.5 124.1 41.1 48.1 41.6 11.6
Et2 ImS 178.74 – – – – – – – – – 46.13 42.69 11.92 –
(5) 177.1 182.1 145.3 133.5 123.7 130.4 28.3 24.0 23.4 124.6 45.9 42.2 11.5 _

shown in Figs. 2, 3 and 4 respectively, whereas PF6 − and C2 H6 O linear coordination geometry owing bond angles of 177.1(1),
groups are removed for clarity. Some particular bond angles [°] 172.9(1) and 176.6(1)° (C—Au—S) for (1), (3) and (4) respectively.
and bond distances [Å] are also enlisted in Table 5. The crystal The close values of bonds suggest isostructural nature of (1)
structures showing [Au(IPr)(thiones)]+ and PF6 − ions confirmed and (4). The bond distances between Au—C and Au—S in (1)
that crystals comprised ionic species connected by electrostatic and (4) are in agreement with literature of similar complexes
interactions. The cationic part bearing gold(I) ion showed almost [74–77]. ligands. In particular, the Au—C bond lengths are 2.003(4),

4
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

pare the cytotoxic potential of these complexes. The concentrations


of complexes (1–5) were precisely and gradually raised along with
the cisplatin standard while keeping the number of cells fixed to
measure the dose-dependent activity of complexes. The IC50 val-
ues (μM) thus obtained are summarized in Fig. 5 and Table 6. The
studies indicated that attachment of thione ligands had no positive
influence on the antiproliferation properties of gold(I)-NHC thione
complexes. Therefore, all synthesized complexes possess low effec-
tiveness than cisplatin in terms of antiproliferative properties. The
lower cytotoxic potential of complexes may resulted from steric
hindrance arising from binding of IPr and thiones. Nevertheless,
the results were overall similar or in some cases better than the
platinum(II) complexes of thiones [78]. The results are also com-
parable with the previous reports where selenone was used as a
co-ligand for gold(I)-NHC complexes [61].

3.4. Interaction of complexes with biomolecules

For designing effective pharmaceutical drugs, the mechanism of


drug-cell interaction has a great significant interest. For instance,
several methods have been used to evaluate the interaction of
Fig. 1. Schematic representation of metal NHC complexes.
the drug with the cell components, such as mass spectrometry,
UV spectrophotometry, and fluorescence spectrometry. The elec-
1.996(3) and 1.982(4) Å for (1), (3) and (4). The bond connection trochemical investigation is another powerful tool to study the
around the sulfur atom is V-shaped with (Au−S−C) bond angles interaction between biomolecules and drugs. For instance, DNA-
103.05(16), 115.12(11), and 102.14(16)° in complexes (1), (3) and modified electrodes have been frequently utilized to investigate the
(4) respectively. A trigonal planar geometry was observed by bond electrochemical interaction between drug molecules and DNA [79].
angles surrounding >C=S and carbene C atoms. The absense of Electrochemistry is helpful to extract the thermodynamic and the
aurophilic interactions in (1), (3) and (4) could result from bulky kinetic information of the drug [80].
groups of the IPr and thiones ligands as in the case of Au(IPr)- Moreover, the drug-protein interaction has been investigated
based selenones [76]. A summary of crystal of above mentioned where the binding number and association constant between drug
complexes is given in Table 4. and protein was determined by electrochemical methods [81]. The
assessment of the interaction between small molecules and pro-
3.3. In vitro anticancer properties teins not only helps elaborate the complex biological mechanisms
but is also one of the most significant phases of drug discov-
The complexes (1–5) were investigated for in vitro cytotoxic ac- ery. Disposable electrochemical cells were used to examine small-
tivities against HCT-15, A549 and MCF7 cancer cells. Cisplatin, a molecule-protein interaction [82]. The tyrosine-derived nanosphere
classical chemotherapy agent, was employed as a reference to com- has been used to investigate their interaction with the drugs, the

Fig. 2. X-ray structure of complex (1).

5
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

Fig. 3. X-ray structure of Complex (3).

Fig. 4. X-ray structure of Complex (4).

most prominent interaction was π −π interactions and hydrogen The interaction of the synthesized gold(I)-NHC thione com-
bonding [83]. The proteins are being used to analyze the electro- plexes (1–5) with model amino acid was investigated. As all these
chemical drug resistance in cancer cells. Several electrochemical complexes were found electrically inactive, a highly electroactive
approaches, including CV and impedance spectroscopy, have been amino acid L-tyrosine was chosen to observe the interaction of the
employed for electrochemical investigation of the drugs [84]. In complexes. Electrochemical investigation of these complexes was
this work, we have used CV and SWV to investigate the interac- done by using CV and SWV, both of which revealed sufficient in-
tion of the complexes with L-tyrosine. formation about complexes interaction with the L- tyrosine by de-

6
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

Table 4
Summary of XRD data for complexes (1), (3) and (4).

Parameters Complex (1) Complex (3) Complex (4)

Empirical formula C30 H42 AuN4 S·PF6 C32 H46 AuN4 S·PF6 ·C2 H6 O C32 H46 AuN4 S·PF6
Mw 832.68 906.79 860.72
Lattice system, Space group Monoclinic, P21 /c Triclinic, P Monoclinic, P21 /c
Temperature (K) 173 173 173
a, b, c [Å] 13.1809(7), 16.4724(7), 15.9454(9) 8.9194(5), 13.5483(7), 16.2561(9) 13.2508(7), 17.4343(7), 16.0079(7)
β [°] 102.399(4) 97.657(4) 102.053(4)
V (Å3 ) 3381.3(3) 1926.85(18) 3616.6(3)
Z 4 2 4
Radiation type Mo Kα Mo Kα Mo Kα
μ (mm−1 ) 4.52 3.97 4.23
Crystal size (mm) 0.45 × 0.32 × 0.19 0.41 × 0.32 × 0.20 0.40 × 0.33 × 0.20
Instrument STOE IPDS 2 diffractometer STOE IPDS 2 diffractometer STOE IPDS 2 diffractometer
Absorption correction Multi-scan(MULABS; Spek, 2009) Multi-scan(MULABS; Spek, 2009) Multi-scan(MULABS; Spek, 2009)
Tmin , Tmax 0.526, 1.000 0.573, 1.000 0.660, 1.000
No. of measured, independent and 46,000, 6393, 4675 26,727, 7266, 6345 49,233, 6847, 4996
observed [I > 2σ (I)] reflections
Rint 0.081 0.052 0.093
(sin θ /λ)max (Å−1) 0.610 0.609 0.610
R[F2 > 2σ (F2 )], wR(F2 ), S 0.026, 0.053, 0.86 0.022, 0.048, 0.94 0.030, 0.064, 0.86
No. of reflections 6393 7266 6847
No. of parameters 432 476 479
No. of restraints 2 6 1

ρ max,
ρ min (e Å−3) 0.99, −0.82 0.94, −1.50 1.70, −0.97

Fig. 5. IC50 values of complexes (1–5) and cisplatin.

creasing the current and peak shift of the L-tyrosine after interac- sponses of 0.5 mM L-tyrosine, as shown in Fig. 6(F, F’), almost re-
tion with complexes. The oxidation peak potential for 0.5 mM L- tained the same peak current intensity and peak potential even af-
tyrosine solution (0.1 M PB, pH 7.0) appeared at 0.598 and 0.620 V ter spiking the same volume of the reagent solvent was used for
for SWV and CV. The high-level interaction of the L-tyrosine was the preparation of the anticancer complexes. The controlled ex-
observed with complex (1), (2) and (4). The maximum interac- periment further confirmed the interaction of synthesized com-
tion was observed with complex (1). The current was decreased plexes and the L-tyrosine. Further investigation of the complexes
sharply and a continuous peak shift was observed from 0.620 to was done by studying the effect of these complexes on the diffu-
0.674 V using cyclic voltammetry for 0.5 mM L-tyrosine by succes- sion coefficient of the L-tyrosine. The scan rate was varied from
sive addition of the complex (1) from 0 μM to 90 μM and simi- 20 to 120 mVs-1 to consider the scan rate effect for 0.5 mM L-
lar trends was observed for SWV (Fig. 6A, A’). Similarly, for com- tyrosine in the absence or presence of 100 μM complexes. This ex-
plexes (2) and (4). The interaction of the drug with L-tyrosine was periment indicated that the raising scan rate increased the current.
observed a little less compared to complex (1) and the peak shift Linear relations have been observed between the square root of
(CVs) was observed almost the same for two complexes from 0.620 scan rate and the oxidation peak current of the L-tyrosine with or
to 0.660 V (Fig. 6B, B’ and D, D’). The complexes (3) and (5) have without complexes. Linear equations yielded for 0.5 mM L-tyrosine
shown very weak interaction with L-tyrosine. A small decline in with or without 100 μM complex (1) were y = 31.784x+0.128 and
current and peak shift was observed for complexes (3) and (5) y = 23.364x+1.068, respectively (Fig. 7). The diffusion coefficients
from 0.620 to 0.629 V and 0.620 to 0.638 V, respectively (Fig. 6C, were found by using Randles–Sevcik equation
C’ and E, E’). As a controlled experiment, the CV and SWV re-
Ip = 2.69 × 105C γ 1/2 D1/2 n3/2 A (1)

7
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

Fig. 6. Cyclic and square wave voltammograms of the gold(I)-NHC thione complexes (1) (A, A ), (2) (B, B ), (3) (C, C ), (4) (D, D ), and (5) (E, E ) in phosphate buffer (pH
7.0, 0.1 M) containing 0.5 mM L-tyrosine at various concentrations of the gold(I) complexes (a) blank, and with concentrations of (b) 0, (c) 10, (d) 30, (e) 50, (f) 60, and (g)
90 μM. The effect on 0.5 mM L-tyrosine solution (F, F ) by adding solvent blank in concentrations of (b) 0, (c) 15, (d) 45, (e) 95, (f) 110 and (g) 155 μL.

In Eq. (1), γ represents the scan rate (V/s), C is the concentration 3.304 × 10−6 for 0.5 mM L-tyrosine compared to the diffusion co-
of the L-tyrosine (mol/L), D is diffusion coefficient (cm2 /s), n rep- efficient in the presence of 100 μM complexes. The complexes (1),
resents the number of electrons, A describes the surface area of (2) and (4) have shown a significant effect on the diffusion co-
the electrode (cm2 ) and Ip is the current (A). In the absence of efficient of the L-tyrosine and it was decreased to 1.786 × 10−6 ,
gold(I) complexes, the diffusion coefficient has a greater value of 2.138 × 10−6 and 2.425 × 10−6 cm2 /s, respectively. The gold(I)

8
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

Fig. 7. Cyclic Voltammograms (A and B) obtained from solution comprising (0.5 mM) L-tyrosine in the absence of (a) and presence of (b) (100 μM) complex (1) using GCE
at scan rates of (a) 20 mV/s (b) 40 mV/s, (c) 60 mV/s, (d) 80 mV/s, (e) 100 mV/s, (f) 120 mV/s. The graphical representation (C) has shown the linear relationship between
current and the square root of the scan rates for (0.5 mM) L-tyrosine in the absence (a) and the presence (b) of (100 μM) complex (1).

Table 5
Selected bond length [Å] and bond angles [°] of (1), (3) and (4).

(1) (3) (4)

Bond length [Å]


Au1−C4 2.003(4) – –
Au1—C6 – 1.996(3) 1.982(4)
Au1—S1 2.299(1) 2.295(1) 2.298(1)
S1—C1 1.707(4) 1.715(3) 1.710(5)
N1—Cl 1.315(6) 1.320(4) 1.318(6)
N1—C2 1.461(6) 1.469(5) 1.471(7)
Bond angles [°]
C4—Au1—S1 177.1(1) – –
C6—Au1—S1 – 172.9(1) 176.6(1)
complexes (3) and (5) also showed an effect on the diffusion coef- C1—S1—Au1 103.1(2) 115.1(1) 102.1(2)
N3—C4—Au1 124.8(3) – –
ficient of L-tyrosine but that effect was not so prominent compared
N3—C6—Au1 – 128.3(2) 131.3(3)
to the rest of the gold(I) complexes (Table 7). N3—C6—N4 105.5(2) 104.2(3) –
The electrochemical study has revealed how the gold(I)-NHC N3—C4—N4 104.8(3) – –
thione complexes affect peak current, oxidation peak potential N1—C1— N2 – 110.1(3) 110.4(4)
and the diffusion coefficient of the L-tyrosine. These substantial N1—C1—S1 127.9(3) 130.3(3) 123.0(4)
N2—C1—S1 122.0(4) 119.5(2) 126.7(4)
changes have confirmed the interaction between L-tyrosine and C1—N1—C2 112.6(4) 111.4(3) 111.0(4)
gold(I)-NHC thione complexes, especially complexes (1), (2) and N4—C6—N3 – – 104.0(4)
(4). This considerable interaction could be attributed to the struc-
ture of the complexes. These three complexes have a secondary
amino group in their structure, thus allowing the electrostatic in-
teraction (hydrogen bonding) between these complexes and the L-
tyrosine through the hydrogen of the secondary amino group. The and the diffusion coefficient was observed. These complexes have
diffusion of L-tyrosine was decreased due to the attachment of tertiary amino groups allowing a weak interaction and further hin-
the bulky gold(I)-NHC thione complex. This effect was found more drance may be created by the presence of methyl in complex (3)
prominent for complex (1) and this effect resulted from the pres- and the ethyl in complex (5) instead of hydrogen. This behavior of
ence of two secondary amino groups in the complexes. The inter- the complex (3) and complex (5) further supported the evidence
action of the complexes (3) and (5) was very weak with L-tyrosine that amino groups facilitate the enhancement of the interaction
and a small decrease in current, peak shift (Fig. 6 C, C’ and E, E’) with the L-tyrosine.

9
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

Table 6 Emeritus Helen Stoeckli-Evans, University of Neuchâtel, Switzer-


IC50 values (μM) of complexes (1–5) and cisplatin against carcinoma cell lines.
land for X-ray diffraction analysis.
Compound HCT-15 A549 MCF7
IC50 values Supplementary materials
(1) 90.68 ± 1.84 102.17 ± 2.57 92.8 ± 0.34
(2) 88.08 ± 0.91 89.98 ± 2.41 94.93 ± 1.68 X-ray crystal data of 3, 4 and 1 have been submitted to the
(3) 74.26 ± 0.59 90.25 ± 1.78 79.76 ± 1.9
Cambridge Crystallographic Data Center via the CCDC Number
(4) 93.90 ± 1.58 83.36 ± 0.71 91.33 ± 2.07
(5) 90.92 ± 1.91 77.90 ± 1.58 71.90 ± 1.58 2,090,806–2,090,808. The data files can be accessed free of cost
Cisplatin 32.04 ± 2.12 42.2 ± 2.01 23.25 ± 3.79 with application at CCDC, 12 Union Road, Cambridge CB2 1EZ, UK,
e-mail: deposit@ccdc.cam.ac.uk or www.ccdc.cam.ac.uk.
Table 7 Supplementary material associated with this article can be
Comparison of the diffusion coefficient of the L-tyrosine in the absence and pres- found, in the online version, at doi:10.1016/j.molstruc.2022.132482.
ence of 100 μM synthesized complexes (1–5).

Complex Concentration of Concentration of Diffusion


References
complex (μM) L-tyrosine (μM) coefficient (cm2 /s)
[1] S. Rafique, et al., Transition metal complexes as potential therapeutic agents,
No 0 500 3.304 × 10−6 Biotechnol. Mol. Biol. Rev. 5 (2) (2010) 38–45.
Complex [2] S.K. Nandanwar, H.J. Kim, Anticancer and antibacterial activity of transition
(1) 100 500 1.786 × 10−6 metal complexes, ChemistrySelect 4 (5) (2019) 1706–1721.
(2) 100 500 2.138 × 10−6 [3] N.V. Loginova, et al., Metal complexes as promising agents for biomedical ap-
(3) 100 500 2.471 × 10−6 plications, Curr. Med. Chem. 27 (31) (2020) 5213–5249.
(4) 100 500 2.425 × 10−6 [4] S. Dasari, P.B. Tchounwou, Cisplatin in cancer therapy: molecular mechanisms
(5) 100 500 2.494 × 10−6 of action, Eur. J. Pharmacol. 740 (2014) 364–378.
[5] A.M. Florea, D. Büsselberg, Cisplatin as an anti-tumor drug: cellular mecha-
nisms of activity, drug resistance and induced side effects, Cancers 3 (1) (2011)
1351–1371.
4. Conclusion [6] R. Oun, Y.E. Moussa, N.J. Wheate, The side effects of platinum-based
chemotherapy drugs: a review for chemists, Dalton Trans. 47 (19) (2018)
6645–6653.
In conclusion, five stable gold(I)-NHC thione complexes (1– [7] M.Y. Wani, M.A. Malik, in: Non-platinum Anticancer Agents, in Gold and Its
5) were synthesized and characterized using advanced analytical Complexes in Anticancer Chemotherapy, Springer, 2021, pp. 51–68.
[8] C.I. Yeo, K.K. Ooi, E.R. Tiekink, Gold-based medicine: a paradigm shift in anti–
techniques. The XRD studies confirmed a nearly linear geometry cancer therapy? Molecules 23 (6) (2018) 1410.
of the gold(I) atom in complexes (1), (3) and (4). The cytotoxicity [9] M.-L. Teyssot, et al., Metal-NHC complexes: a survey of anti-cancer properties,
data showed that thione ligands did not contribute to the cytotoxic Dalton Trans. (35) (2009) 6894–6902.
[10] J.D. Knoll, C. Turro, Control and utilization of ruthenium and rhodium metal
potential of gold(I)-NHC complexes. Hence, the synthesized com- complex excited states for photoactivated cancer therapy, Coord. Chem. Rev.
plexes have a low cytotoxic effect than cisplatin and a weak ability 282 (2015) 110–126.
to control cancer proliferation. When compared with platinum(II) [11] D. Denoyer, et al., Targeting copper in cancer therapy:‘copper that cancer’,
Metallomics 7 (11) (2015) 1459–1476.
thione based complexes, the results show similar or slightly bet-
[12] C.P. Tan, et al., Metallomics insights into the programmed cell death induced
ter performance in some instances. The study also provides an op- by metal-based anticancer compounds, Metallomics 6 (5) (2014) 978–995.
portunity to compare the anticancer potential of thiones and se- [13] B. Englinger, et al., Metal drugs and the anticancer immune response, Chem.
lenones, indicating that selenium imparts more anticancer charac- Rev. 119 (2) (2018) 1519–1624.
[14] P. Jia, et al., Recent advances and future development of metal complexes as
ter to the complexes than thiones. anticancer agents, J. Coord. Chem. 70 (13) (2017) 2175–2201.
The electrochemical investigations have concluded the interac- [15] M. Hanif, C.G. Hartinger, Anticancer metallodrugs: where is the next cisplatin?
tion of the anticancer drug molecules with the L-tyrosine which is Future Med. Chem. 10 (6) (2018) 615–617.
[16] A. Garoufis, S. Hadjikakou, N. Hadjiliadis, Palladium coordination compounds
a vital component of the proteins. All five complexes have shown as anti-viral, anti-fungal, anti-microbial and anti-tumor agents, Coord. Chem.
interaction with the L-tyrosine which was observed by the de- Rev. 253 (9-10) (2009) 1384–1397.
crease of the oxidation peak current, change in oxidation peak po- [17] S.P. Pricker, Medical uses of gold compounds: past, present and future, Gold
Bull. 29 (2) (1996) 53–60.
tential and by decreasing the diffusion coefficient of the L-tyrosine. [18] C. Bombardier, et al., Auranofin therapy and quality of life in patients with
The possible interaction may be taking place through a secondary rheumatoid arthritis. Results of a multicenter trial, Am. J. Med. 81 (4) (1986)
amino group and weak interaction through a tertiary amino group. 565–578.
[19] A.E. Finkelstein, et al., Auranofin. New oral gold compound for treatment of
rheumatoid arthritis, Ann. Rheum. Dis. 35 (3) (1976) 251.
Declaration of Competing Interest [20] S. Yue, et al., Recent advances of gold compounds in anticancer immunity,
Front. Chem. 8 (2020) 543.
There are no conflicts to declare. [21] A. Casini, et al., Gold (III) compounds as anticancer agents: Relevance of
gold–protein interactions for their mechanism of action, J. Inorg. Biochem. 102
(3) (2008) 564–575.
CRediT authorship contribution statement [22] L. Messori, et al., The mode of action of anticancer gold-based drugs: a struc-
tural perspective, Chem. Commun. 49 (86) (2013) 10100–10102.
[23] A. Casini, et al., Gold(III) compounds as anticancer agents: Relevance of
Sughra Gulzar: Conceptualization. Umme Ammara: Methodol-
gold–protein interactions for their mechanism of action, J. Inorg. Biochem. 102
ogy. Zeeshan Abid: Methodology. Munazza Shahid: Formal anal- (3) (2008) 564–575.
ysis. Raja Shahid Ashraf: Methodology. Nadeem Baig: Data cura- [24] A. Bindoli, et al., Thioredoxin reductase: a target for gold compounds acting as
tion. Abdel-Nasser Kawde: Data curation. Gaurav Bhatia: Formal potential anticancer drugs, Coord. Chem. Rev. 253 (11) (2009) 1692–1707.
[25] P. Zhang, P.J. Sadler, Advances in the design of organometallic anticancer com-
analysis. Anvarhusein A. Isab: Validation. Muhammad Altaf: Su- plexes, J. Organomet. Chem. 839 (2017) 5–14.
pervision. [26] A.L. Lainé, C. Passirani, Novel metal-based anticancer drugs: a new challenge
in drug delivery, Curr. Opin. Pharmacol. 12 (4) (2012) 420–426.
[27] C.K. Adokoh, Therapeutic potential of dithiocarbamate supported gold com-
Acknowledgement
pounds, RSC Adv. 10 (5) (2020) 2975–2988.
[28] M. Altaf, et al., New bipyridine gold(III) dithiocarbamate-containing complexes
The authors express their gratitude to King Fahd University of exerted a potent anticancer activity against cisplatin-resistant cancer cells in-
Petroleum and Minerals, Interdisciplinary Research Center for Ad- dependent of p53 status, Oncotarget 8 (1) (2017) 490–505.
[29] M. Altaf, et al., Potent in vitro and in vivo anticancer activity of new bipyridine
vanced Materials and greatly appreciate the financial support un- and bipyrimidine gold (III) dithiocarbamate derivatives, Cancers 11 (4) (2019)
der the project No. INAM2102. We are also thankful to Professor 474.

10
S. Gulzar, U. Ammara, Z. Abid et al. Journal of Molecular Structure 1255 (2022) 132482

[30] A.A. Sulaiman, et al., Synthesis, characterization, DFT optimization and anti- [58] M. Mora, M.C. Gimeno, R. Visbal, Recent advances in gold–NHC complexes
cancer evaluation of phosphanegold(I) dithiocarbamates, J. Mol. Struct. 1218 with biological properties, Chem. Soc. Rev. 48 (2) (2019) 447–462.
(2020) 128486. [59] N. Razzaghi-Asl, et al., Insights into the current status of privileged N-hetero-
[31] M. Altaf, et al., Synthesis, characterization and anticancer activity of gold(I) cycles as antileishmanial agents, Mol. Divers. 24 (2) (2020) 525–569.
complexes that contain tri-tert-butylphosphine and dialkyl dithiocarbamate [60] M.G. Karaaslan, et al., Chemistry, structure, and biological roles of Au-NHC
ligands, Eur. J. Med. Chem. 95 (2015) 464–472. complexes as TrxR inhibitors, Bioorg. Chem. 95 (2020) 103552.
[32] A.A.A. Sulaiman, et al., Synthesis, characterization, and in vitro cytotoxicity of [61] A.A.A. Seliman, et al., Synthesis, X-ray structures and anticancer activity of
gold(i) complexes of 2-(diphenylphosphanyl)ethylamine and dithiocarbamates, gold(I)-carbene complexes with selenones as co-ligands and their molecular
Z. Anorg, Allg. Chem. 642 (24) (2016) 1454–1459. docking studies with thioredoxin reductase, J. Organomet. Chem. 848 (2017)
[33] M. Altaf, et al., The synthesis, spectroscopic characterization and anticancer 175–183.
activity of new mono and binuclear phosphanegold(i) dithiocarbamate com- [62] A.A.A. Seliman, et al., Synthesis, X-ray structure and cytotoxicity evaluation of
plexes, New J. Chem. 39 (1) (2015) 377–385. carbene-based gold(I) complexes of selenones, Inorg. Chim. Acta 476 (2018)
[34] S.S. Al-Jaroudi, et al., Synthesis, characterization, in vitro cytotoxicity and DNA 46–53.
interaction study of phosphanegold(I) complexes with dithiocarbamate ligands, [63] B. Dominelli, J.D.G. Correia, F.E. Kühn, Medicinal applications of
Inorg. Chim. Acta 464 (2017) 37–48. gold(I/III)-based complexes bearing N-heterocyclic carbene and phosphine
[35] Z. Faghih, et al., Synthesis, molecular docking and cytotoxic activity evaluation ligands, J. Organomet. Chem. 866 (2018) 153–164.
of organometallic thiolated gold (I) complexes, Iran. J. Pharm. Sci. 19 (3) (2020) [64] F. Guarra, et al., Cytotoxic Ag(I) and Au(I) NHC-carbenes bind DNA and show
134. TrxR inhibition, J. Inorg. Biochem. 205 (2020) 110998.
[36] L. Rouco, et al., Combined effect of caspase-dependent and caspase-indepen- [65] S. Huang, et al., Synthesis and in vitro anticancer activities of selenium N-hete-
dent apoptosis in the anticancer activity of gold complexes with phosphine rocyclic carbene compounds, Chem. Biol. Drug Des. 98 (3) (2021) 435–444.
and benzimidazole derivatives, Pharmaceuticals 14 (1) (2021) 10. [66] M. Altaf, et al., Synthesis, X-ray structures, spectroscopic analysis and anti-
[37] H.V. Le, et al., Highly cytotoxic gold(i)-phosphane dithiocarbamate complexes cancer activity of novel gold(I) carbene complexes, J. Organomet. Chem. 765
trigger an ER stress-dependent immune response in ovarian cancer cells, Dal- (2014) 68–79.
ton Trans. 49 (22) (2020) 7355–7363. [67] B.V. Trzhtsinskaya, N.D. Abramova, Imidazole-2-thiones: synthesis, structure,
[38] J.O. Adeyemi, D.C. Onwudiwe, The mechanisms of action involving dithiocar- properties, Sulfur Rep. 10 (4) (1991) 389–421.
bamate complexes in biological systems, Inorg. Chim. Acta 511 (2020) 119809. [68] J.K. Savjani, A.K. Gajjar, Pharmaceutical Importance and synthetic strategies for
[39] E. Abas, et al., New selective thiolate gold (i) complexes inhibit the prolifera- imidazolidine-2-thione and, Pak. J. Biol. Sci. 14 (24) (2011) 1076–1089.
tion of different human cancer cells and induce apoptosis in primary cultures [69] F. Caddeo, et al., Gold thione complexes, Inorganics 2 (3) (2014) 424–432.
of mouse colon tumors, Dalton Trans. 49 (6) (2020) 1915–1927. [70] G. Kirishnamaline, et al., Theoretical investigation of structure, anticancer ac-
[40] B.M. Sutton, Gold compounds for rheumatoid arthritis, Gold Bull. 19 (1) (1986) tivity and molecular docking of thiourea derivatives, J. Mol. Struct. 1225 (2021)
15–16. 129118.
[41] V.Z. Mota, et al., Gold complexes with benzimidazole derivatives: synthesis, [71] W.A. Souza, et al., Crystal structure and spectroscopy properties of new PtII
characterization and biological studies, Biometals 27 (1) (2014) 183–194. complexes containing 5-alkyl-1, 3, 4-oxadiazol-2-thione derivatives, J. Mol.
[42] C. Zhang, et al., Synthesis, characterization, and antileishmanial activity of neu- Struct. 1226 (2021) 129250.
tral N-heterocyclic carbenes gold(I) complexes, Eur. J. Med. Chem. 143 (2018) [72] A.A. Seliman, et al., Synthesis, X-ray structure and cytotoxicity evaluation of
1635–1643. carbene-based gold (I) complexes of selenones, Inorg. Chim. Acta 476 (2018)
[43] F. Nareetsile, et al., Transition metal complexes with HIV/AIDS inhibitory prop- 46–53.
erties, Chem. Rev. Lett. 3 (3) (2020) 140–160. [73] S. Ftouh, et al., Synthesis, characterization, and antileishmanial activity of neu-
[44] P.N. Fonteh, F.K. Keter, D. Meyer, HIV therapeutic possibilities of gold com- tral gold (I) complexes with N-heterocyclic carbene ligands bearing sulfur-con-
pounds, Biometals 23 (2) (2010) 185–196. taining side arms, Organometallics 40 (10) (2021) 1466–1473.
[45] M. Gaba, C. Mohan, Development of drugs based on imidazole and benzimida- [74] A.A. Seliman, et al., Synthesis, X-ray structures and anticancer activity of
zole bioactive heterocycles: recent advances and future directions, Med. Chem. gold (I)-carbene complexes with selenones as co-ligands and their molecular
Res. 25 (2) (2015) 173–210. docking studies with thioredoxin reductase, J. Organomet. Chem. 848 (2017)
[46] Y. Bansal, O. Silakari, The therapeutic journey of benzimidazoles: a review, 175–183.
Bioorg. Med. Chem. 20 (21) (2012) 6208–6236. [75] S. Huang, et al., Synthesis and in vitro anticancer activities of selenium N-hete-
[47] D.K. Agarwal, P.K. Goyal, Synthetic and biological aspects of benzimidazole rocyclic carbene compounds, Chem. Biol. Drug Des. 98 (3) (2021) 435–444.
derivatives, Chem. Biol. Interface 11 (1) (2021) 6–12. [76] B. Yu, et al., Synthesis, characterization, and antitumor properties of Au
[48] P. Sharma, et al., Imidazoles as potential anticancer agents: an update on re- (I)–thiourea complexes, Metallomics 12 (1) (2020) 104–113.
cent studies, Molecules 26 (14) (2021) 4213. [77] Y.Q. Zhao, et al., New gold (I) complexes with 5-aromatic ring-1, 3, 4-oxadi-
[49] X. Liu, et al., Imidazole and benzimidazole modified half-sandwich iridiumII- azole-2-thione and triphenylphosphine as potential multifunctional materials,
IN-heterocyclic carbene complexes: synthesis, anticancer application, and or- Tetrahedron Lett. 61 (52) (2020) 152668.
ganelle targeting, Front. Chem. 8 (2020) 182. [78] A.Z.A. Mustafa, et al., Tetrakis(thione)platinum(II) complexes: synthesis, spec-
[50] K.C. Howard, et al., A comprehensive overview of the medicinal chemistry troscopic characterization, crystal structures, and in vitro cytotoxicity, J. Coord.
of antifungal drugs: perspectives and promise, Chem. Soc. Rev. 49 (8) (2020) Chem. 68 (19) (2015) 3511–3524.
2426–2480. [79] S. Rauf, et al., Electrochemical approach of anticancer drugs–DNA interaction,
[51] S.S. Alghamdi, et al., Imidazole as a promising medicinal scaffold: current sta- J. Pharm. Biomed. Anal. 37 (2) (2005) 205–217.
tus and future direction, Drug Des. Dev. Ther. 15 (2021) 3289–3312. [80] E.A. Hillard, et al., Electrochemical parameters and techniques in drug develop-
[52] F. Guarra, et al., A focus on the biological targets for coinage metal-NHCs as ment, with an emphasis on quinones and related compounds, Chem. Commun.
potential anticancer complexes, J. Inorg. Biochem. 217 (2021) 111355. (23) (2008) 2612–2628.
[53] D. Iacopetta, et al., Is the way to fight cancer paved with gold? Metal-based [81] Y. Wu, X. Ji, S. Hu, Studies on electrochemical oxidation of azithromycin and
carbene complexes with multiple and fascinating biological features, Pharma- its interaction with bovine serum albumin, Bioelectrochemistry 64 (1) (2004)
ceuticals 13 (5) (2020) 91. 91–97.
[54] I. Slimani, et al., Novel N-heterocyclic carbene silver(I) complexes: synthesis, [82] C.V. Uliana, et al., Label-free evaluation of small-molecule–protein interaction
structural characterization, antimicrobial and cytotoxicity potential studies, J. using magnetic capture and electrochemical detection, Anal. Bioanal.Chem. 411
Braz. Chem. Soc. 31 (2020) 2058–2070. (10) (2019) 2111–2119.
[55] M. Koy, et al., N-Heterocyclic carbenes as tunable ligands for catalytic metal [83] A.D. Costache, et al., Polymer−drug interactions in tyrosine-derived triblock
surfaces, Nat. Catal. 4 (5) (2021) 352–363. copolymer nanospheres: a computational modeling approach, Mol. Pharm. 6
[56] L. Oehninger, R. Rubbiani, I. Ott, N-Heterocyclic carbene metal complexes in (5) (2009) 1620–1627.
medicinal chemistry, Dalton Trans. 42 (10) (2013) 3269–3284. [84] R. Islam, H.T.L. Luu, S. Kuss, Electrochemical approaches and advances towards
[57] M.N. Hopkinson, et al., An overview of N-heterocyclic carbenes, Nature 510 the detection of drug resistance, J. Electrochem. Soc. 167 (4) (2020) 045501.
(7506) (2014) 485–496.

11

You might also like