You are on page 1of 20

Coordination Chemistry Reviews 436 (2021) 213849

Contents lists available at ScienceDirect

Coordination Chemistry Reviews


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

Review

Biological properties of ruthenium(II)/(III) complexes with flavonoids


as ligands
Magdalena Małecka a,⇑, Anna Skoczyńska b,c, David M. Goodman d, Christian G. Hartinger d,
Elzbieta Budzisz b,⇑
a
Department of Physical Chemistry, Faculty of Chemistry University of Lodz, Pomorska 163/165, 90-236 Łódź, Poland
b
Cosmetic Row Materials Chemistry, Faculty of Pharmacy, Medical University of Łódź, Muszynskiego 1, 91-419, Poland
c
Department of Pharmacology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellonska 4, 41-200 Sosnowiec, Poland
d
University of Auckland, School of Chemical Sciences, Private Bag 92019, Auckland 1142, New Zealand

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

Article history: This review highlights the progress in the development of ruthenium(II)/(III) complexes with flavone
Received 4 November 2020 derivatives as potential therapeutic agents. We focused on natural hydroxyflavone derivatives and their
Received in revised form 3 February 2021 synthetic amino analogues as ligands in ruthenium(II)/(III) complexes which demonstrate antimicrobial,
Accepted 4 February 2021
antitumor activity and/or enzyme inhibition. Some enzymes targeted by flavone derivatives are impor-
Available online 8 March 2021
tant for the development and progression of cancer cells which underpins the design concept for the
Ru complexes as multitargeted compounds consisting of a bioactive ligand and a ruthenium center.
Keywords:
Ó 2021 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://
Flavonoids
Ruthenium(II)/(III) complexes
creativecommons.org/licenses/by/4.0/).
Antitumor activity

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
2. Therapeutic properties of flavonoids. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Biological properties of Ru(II)/(III) complexes with flavonoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1. Ruthenium compounds of O-donor flavonoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.1.1. Ruthenium coordination compounds – From mono- to dinuclear anticancer agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.1.2. Bioorganoruthenium chemistry: Ru(g6-arene) complexes of O-donor flavonoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
3.2. Ru(II)/(III) complexes of aminoflavonoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Appendix A. Supplementary data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15

1. Introduction complexes containing chromone, coumarin and flavonoid ligands


have demonstrable activity against leukemia, lung, cervical, and
For several years our scientific efforts have been focused on melanoma cancer cell lines [2–5] as well as being efficacious
metal complexes containing flavone-derived ligands and exploit- against Huntington’s [6–8], b-thalassemia [9–11], heart diseases
ing their interesting chemical and biological properties [1]. Metal and diabetes mellitus [12–15]. Plant-synthesized flavones [16]
are biologically interesting molecules, due to the structure-
⇑ Corresponding authors. dependent medicinal and biological properties [17], which include
E-mail addresses: magdalena.malecka@chemia.uni.lodz.pl (M. Małecka), antioxidant [18], antimicrobial [19], antiviral [20], antibacterial
elzbieta.budzisz@umed.lodz.pl (E. Budzisz). [20], antidiabetic, and anti-inflammatory activity, in addition to

https://doi.org/10.1016/j.ccr.2021.213849
0010-8545/Ó 2021 The Authors. Published by Elsevier B.V.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

metal chelation [16]. Flavones are polyphenolic compounds with a (transforming growth factor beta) and bFGF (basic fibroblast
general structure consisting of two benzene rings (A and B) and a growth factor). Isoflavone 4 inhibited the growth of breast cancer
heterocyclic pyran ring (C) fused to ring A. This compound class MCF-7 and MDA-MB-468 cells, which are estrogen receptor-
is divided into six subclasses, i.e. flavonols, flavanols, flavones, fla- negative, as well as of estrogen receptor-positive MCF-7-D40 cells
vanones, isoflavones, and anthocyanidins [21], with the differences with IC50 values from 6.5 to 12 mg/ml [18]. Polymethoxylated citrus
between the subclasses arising from the substitution position of flavones, like tangeretin 6 and nobiletin 7, inhibited cell growth in
ring B with C, the saturation and substitution of ring B, and the human squamous cell carcinoma and gliosarcoma cells at concen-
functionalization pattern of the rings through hydroxylation and trations ranging from 5 to 20 mM [43,44] with no activity against
glycosylation [22]. human diploid fibroblast-like lung cells (CC1135) [45]. Tangeretin
The biological activity of Ru complexes has been widely inves- 6 caused apoptosis in HL-60 leukemia cells at concentrations >
tigated. Ruthenium(III) complexes have shown remarkable antitu- 2.7 mM. Additionally, the flavanols, which are tea polyphenols such
mor activity with various advantages over traditional platinum as epicatechin (EC) 8, epicatechin gallate (ECG) 9, epigallocatechin
drugs, including potent efficacy, low toxicity and less drug resis- (EGC) 10, and epigallocatechin gallate (EGCG) 11 [46], suppressed
tance [23–25], while Ru(II) compounds, in particular, organo-Ru human cancer cell growth [47] with EGCG 11 being shown to also
compounds have demonstrated very different biological properties inhibit the metastasis of B16 melanoma cells. These tea polyphe-
including in in vivo studies [26,27]. nols target the Bcl-2 family proteins thus inhibiting their antiapop-
The combination of Ru(II)/(III) complexes with bioactive flavone totic activity. Shukla and Gupta [48] observed that apigenin 12
ligands can act synergistically resulting in superior biological activ- induced apoptosis in prostate cancer cells DU145. (+)-Catechin
ity. Ruthenium(II)-arene complexes with high activity towards 13 can bind to laminin, which is involved in metastasis of malig-
both primary and secondary metastatic tumors have been reported nant tumor cells [49]. Bagli et al. [50] found that fisetin 2, luteolin
[28,29]. The in vivo antioxidant properties of flavonoids may be 3, apigenin 12, 3–hydroxyflavone 14, 30 ,40 -dihydroxyflavone 15,
modulated by metal coordination and thereby affect enzyme inhi- and 20 ,30 -dihydroxyflavone 16 inhibited the in vitro angiogenesis
bitory activity. Similar observations have been made for other of tumor cells. Flavonoids obtained from Trichosanthe kirilowii
bioactive ligand–metal center complexes, resulting in multimodal extract, such as isorhamnetin 17, genkwanin 18 and acacetin 19,
anticancer agents [1,30]. The modes of action of multimodal anti- induced cell cycle arrests, apoptosis, and autophagy in human
cancer agents is influenced by complex stability and as such, Ru breast cancer cells. In addition to PKC and PTK, flavonoids can inhi-
compounds were compared to selected Fe derivatives, due to oxi- bit other kinases including phosphoinositol kinase, phosphatidyli-
dation state and structural similarities [31]. Herein, we summarize nositol kinase, or cyclin-dependent kinase-4 [51,52]. Moreover,
comprehensively the effect of coordination of flavonoid ligands to flavonoids can inhibit transcription factors like NFjB [53], involved
Ru(II) and Ru(III) centers on their biological activities [30]. in the regulation of inflammatory chemokines and cytokines [54]
with additional potential to inhibit cyclooxygenase (COX) and
2. Therapeutic properties of flavonoids lipoxygenase (LOX) enzymes which catalyze the arachidonic acid
metabolism and inhibit inflammatory mediators [55]. Inhibition
Flavonoids are known for their interesting bioactivity profiles of the biosynthesis of such mediators can result from a lower activ-
which make them useful for medical and pharmaceutical applica- ity of enzymes such as PLA2 (phospholipases A2) [56,57], LOX [58]
tions. The development of flavonoids as potential anticancer agents or COX [59]. Furthermore, flavonoids modulate the impact of tran-
is the most widely studied application with further evidence scription factors such as GATA-3 and activate transcription factor 6
demonstrating efficacy as antioxidant, antiinflammatory, antiasth- (STAT-6) [60,61].
matic, antimicrobial and antidiabetic agents. These indications Molecular docking studies can play an important role in drug
have been explored through enzyme inhibition and other protein design [62,63], aiming to predict protein target-ligand binding
interactions such as the molecular docking between quercetin 1 modes and related affinities [64]. Successful docking methods
and inducible nitric oxide synthase (iNOS). explore three-dimensional spaces effectively and the score func-
The anticancer activity of flavonoids arises from the targeting of tion rates potential complementarity to binding sites [65]. Several
kinases, apoptotic proteins, and growth factors thus impacting cell molecular docking studies have been conducted to support inves-
growth, matrix metalloproteinase (MMP) secretion and angiogen- tigations on the targets of flavonoids [66–68], including cyclin-
esis [32]. Some of the enzymes targeted are protein kinase C dependent kinases (CDK) with 8-CF (8-chloroflavone) being docked
(PKC), epidermal growth factor receptor (EGFR), and focal adhesion with CDK6 with binding energies between 13.22 and 9.47 kcal/-
kinase (FAK), which are important for the development and pro- mol and hydrophobic and hydrogen bonding interactions
gression of cancer. Quercetin 1 (Fig. 1), a polyhydroxylated flavo- observed. These results validated the hypothesis of CDK6 inhibi-
noid, is the most active inhibitor of PKC [33–35] with Ferriola tion by 8-CF and supported the experimental results which showed
et al. [36] showing that quercetin 1, fisetin 2 and luteolin 3 inhib- that 8-CF induced cell cycle arrest in the G1 phase caused by CDK6
ited rat brain PKC through blocking the catalytic unit of PKC at the inhibition [69]. Furthermore, Ding et al. [70] used molecular dock-
ATP binding site. Quercetin 1 and luteolin 3 decreased MMP-2 and ing supported by molecular dynamics (MD) simulations to derive
MMP-9 activity in cancer cells such as A549, A431, and MiaPaCa-2 structure–activity relationships for hemoglobin binding by con-
[37]. It was found that quercetin 1 interrupts the elongation of firming the stability of the molecular docking results (entry code
telomeric ends [38]. Luteolin 3 induced apoptosis and caused 2DN1, Fig. 2) [71]. Both methods were in agreement demonstrating
oligonucleosomal DNA cleavage, cytoplasmic blebbing and PARP significant conformational changes upon flavonoid binding except
(poly[ADP-ribose] polymerase 1) degradation in A431 epidermoid for a small rotation of the hydroxyl group in galangin 20, resulting
carcinoma cells and in pancreatic cancer MiaPaCa-2 at 15-25 mM in different non-covalent interactions.
[39]. According to Akiyama et al. [40] isoflavone 4 and genistein In case of the novel aurora kinase inhibitor luteolin 3, molecular
5 are specific inhibitors of protein tyrosine kinase (PTK) and 5 docking studies at the ATP-binding site of aurora kinase (PDB
exhibited antiproliferative activity against HL-60 and K-562 ID:1OL6) [72] indicated stabilization of the inhibitor-protein com-
human cells while 4 suppressed EGFR, and tyrosine kinase activity plex through a bidentate H-bonding pattern between 3 and Ala213
in A431 human cells [41]. Within eight hours, compound 5 induced (Fig. 3) concluding that the introduction of a hydroxyl group at
apoptosis in HL-60 cells [42], due to the inhibition of EGFR, TGF-b position 5 could increase the bioactivity [73].

2
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 1. Structures of compounds listed in the publication.

Studies by Kartasasmita and co-workers [74] investigated inhi- results for iNOS (PDB ID: 1M9T) [75] showed hydrogen bonding
bitory activities of the flavonoids kaempferol 26, quercetin 1, api- between the amide groups of Gly365 and Trp366 and the respec-
genin 12, and primuletin 35 against (iNOS), which catalyzes the tive flavonoids (Fig. 4). These studies demonstrated that the 4-
production of nitric oxide from L-arginine. The molecular docking carbonyl group and the planar fused bicyclic region are essential

3
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 1 (continued)

structural features for the binding. Additionally, there was correla- studies to develop inhibitors of 5-LOX [79]. The studies of Singh
tion between the experimental IC50 values and calculated binding et al. [80] revealed that only six flavonoids of the 26 compounds
energies (DG) (R2 = 0.686). selected were found to dock successfully with 5-LOX with the most
As mentioned above, flavonoids are LOX inhibitors [55] and its potent inhibitors, genkwanin 18, baicalein 22, chrysin 23, galangin
inhibition may be a strategy to prevent carcinogenesis [76–78]. As 20 and velutin 24, being compared to the known 5-LOX inhibitor
secondary metabolites commonly act as negative feedback inhibi- zileuton [81]. This study with further analyses suggested velutin
tors, the secondary metabolites of the human isozyme 5-LOX were 24 as the most promising lead compound for anticancer activity
screened for pharmacokinetic properties by molecular docking through the inhibition of 5-LOX (Fig. 4) [82].

4
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 1 (continued)

Molecular docking studies of the E6 onco-protein, a subcellular inhibit free radical production and scavenge ROS (Reactive Oxygen
target for human papillomavirus (HPV) therapeutics [83], with the Species) and RNS (Reactive Nitrogen Species) [88,89]. Additionally,
herbal flavonoids apigenin 12 and silymarin [84] found that sily- flavonoids inhibit cAMP phosphodiesterase activity responsible for
marin binds through the formation of hydrogen bonds to Tyr32 the regulation of cell function during inflammation by blocking
and Arg55 and is a suitable candidate for further development. cAMP degradation and prolonging cAMP signaling [90,91]. Further-
Another secondary metabolite, i.e., apigenin-7-O-rhamnosyl-6- more, flavonoids inhibit proinflammatory cytokine and chemokine
C-glucoside and its derivatives, obtained from the leaves of Ery- production in neutrophils, mast cells and other immune cells [92–
thrina crista-galli, were investigated by molecular docking [85] 94], in addition to inhibiting or decreasing the activation of
for binding to the human estrogen receptor R (ERR, PDB ID: immune cells involved in inflammation processes such as mono-
1A52) [86]. Interactions to Arg394, Glu353 and His524 were iden- cytes [95,96].
tified to be similar to how 17b-estradiol interacted with the active Asthma is characterized by chronic inflammation of the respira-
site, however, with a higher correlation factor. The lead compound tory tract, and can be improved by flavones, flavonols, anthocyani-
apigenin-7-O-rhamnosyl-6-C-glucoside formed additional hydro- dins, and isoflavones through their anti-inflammatory properties.
gen bonds to amino acids in the pocket. Luteolin 3 caused less prostaglandin D2 and histamine release
Meta-analyses have shown that both individual and total flavo- from human mast cells [97] while tetramethoxyluteolin (methlut)
noid consumption reduce the risk of numerous cancers [87]. This 25 has potential to stop inflammatory mediators from human mast
may be due to the potent antioxidant properties of flavonoids with cells. Quercetin 1, was found to inhibit IL-4 and IL-5 cytokines [98]
these properties being dependent on the structure, specific pattern with kaempferol 26 being shown to block degranulation of eosino-
of substitution and presence of phenolic hydrogens. Flavonoids phils in a murine lung tissue model [60,91,99]. The reduction of

5
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

gets [106]. Administration of quercetin 1 in rats for 14 days


resulted in diminished oxidative stress, reduced inflammation,
improved heart architecture and can relieve or eliminate symp-
toms associated with heat stroke [107]. Similarly, isoflavone 4
and puerarin 28 have been shown to suppress atherosclerosis in
rabbits [108]. It was found in different animal models that the fla-
vone baicalin 22 protected the cardiovascular system by improving
myocardial hypertrophy, cardiac dysfunction, and decreasing
apoptosis in cardiac tissue [109]. Additionally, through the pres-
ence of hydroxyl groups, flavonoids, such as chrysin 23, found in
honey and passionflower, have been shown to inhibit platelet acti-
vation and prevent clot formation [110].
Apigenin 12, galangin 20, isoflavones, flavanones, flavone and
flavonol and other glycosides possess antibacterial activity
[21,111] by inactivating microbial adhesins, enzymes, and trans-
port proteins [112,113]. Robinetin 29, myricetin 30, and ()-
epigallocatechin 31 were shown to inhibit replication of DNA from
Proteus vulgaris with other studies demonstrating that 1, 12 and
Fig. 2. Results of molecular docking calculations for galangin 20 (orange) into
3,6,7,30 ,40 -pentahydroxyflavone 32 affect Escherichia coli DNA gyr-
hemoglobin. The apoprotein is shown as light cyan, and the average position of ase activity [114]. Additional antibacterial properties were found
protein as light pink. The average conformation for flavone is drawn in yellow. for naringenin 33 and sophoraflavanone G 34, which are
Reproduced with permission from ref. [70]. Ó RSC 2016. particularly active towards Streptococci and methicillin-resistant
Staphylococcus aureus (MRSA). According to Osawa et al. [115]
5-hydroxyflavanones 35 and 5-hydroxyisoflavanones substituted
with one, two or three hydroxyl groups at the 7, 20 or 40 positions
stopped the growth of both Streptococcus sobrinus and Streptococ-
cus mutans. Similarly, Haraguchi et al. [116] studied flavonoids
licochalcones A 36 and C 36a, obtained from Glycyrrhiza inflata,
were active against Micrococcus luteus and Staphylococcus aureus.
Many derivatives mirror the antibacterial properties of flavonoids
derived from edible and medicinal plants [117–119].
The antidiabetic properties of flavonoids are due to the induc-
tion of insulin release and activation of pancreatic b-cells [120]
with 11 causing a hypoglycemic effect by inhibition of glucose syn-
thesis in hepatocytes. Kaempferol 26 can reduce oxidative stress in
the endoplasmic reticulum (ER) giving it therapeutic potential
[121,122]. Flavonoids isolated from fermented soybean paste, like
8-hydroxydaidzein 37 and 30 -hydroxydaidzein 38, exhibited a
reduction of melanin synthesis in B16 melanoma cells in compar-
ison to treatment with kojic acid [123]. According to Moon and
Kim [124], hesperidin 39 showed anti-inflammatory activity,
because of decreased formation of proinflammatory cytokines, IL-
Fig. 3. Schematic predicted binding modes of luteolin 3 in the ATP active site of 8 and TNFa, which were induced by hydrogen peroxide in human
aurora kinase (PDB ID 1OL6). keratinocytes (HaCaT cells). The antiulcerogenic activity of api-
genin 12, naringenin 33 and aromadendrin 40, isolated from
Euphorbia cuneata, was demonstrated in the gastric ulcer ethanol
allergic inflammation and lessened release of cytokines was medi- model [125].
ated through mechanisms involving the blockade of NFjB [67]. The
anti-inflammatory properties of genistein 5 are exhibited through
the inhibition of proinflammatory cytokine release from mast cells 3. Biological properties of Ru(II)/(III) complexes with flavonoids
and decreased airway inflammation through the inhibition of tran-
scription factors GATA-3 and STAT-6 [60]. Cyanidin 27, an antho- With the rise of Ru complexes as potentially being complemen-
cyanidin, attenuated inflammation induced by Th17 in a mouse tary to Pt-based anticancer chemotherapy, many Ru(II) and Ru(III)
model for severe asthma [100]. Research conducted on Japanese coordination compounds and Ru(II)-based organometallics have
women proved that eating flavonoid rich soybean can diminish been reported with their anticancer activity assayed both in vitro
the prevalence of allergic rhinitis [101]. and in vivo. Common derivatives are often designed around the
Flavonoids might prevent cardiovascular disease due to their clinically investigated Ru(III) compounds KP1019, KP1339 (IT-
anti-inflammatory, antioxidative and vasodilatory properties by 139/BOLD-100), and NAMI-A and the widely studied organoruthe-
increasing HDL (High Density Lipoprotein) concentration [102]. A nium compound classes of RAED and RAPTA (Fig. 5) [30,126–128].
recent study showed strong evidence that consumption of flavo- Research in this area has shown over the last decade that subtle
noids rich foods corresponded with a diminished risk of mortality changes in the structure often have a significant effect on the phar-
due to cardiovascular diseases [103] with significantly decreased macological properties of metal complexes. To investigate the syn-
cholesterol accumulation due to the impact of 1, indicated by the ergistic activity between flavonoids and bioactive Ru centers, many
presence of quercetin 1 metabolites in rabbit aortas [104]. Querce- complexes have been prepared and studied in biological assays.
tin 1 inhibited the maturation of dendritic cells in mice [105] and Similar effects have been observed for other combinations of bioac-
activated macrophages in inflamed arteries are also potential tar- tive ligands and their metal complexes and therefore has been
6
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 4. Schematic view of interactions between quercetin (1) and iNOS (PDB ID: 1M9T) (left) and of 5-LOX (PDB ID 3V99) with its inhibitor velutin 24 (right).

Fig. 5. Structures of ruthenium(III) compounds investigated in clinical trials and of widely investigated Ru(II) anticancer agents.

Fig. 6. Structures of RuII(polypyridyl)(flavonoid) compounds.

7
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

used as a design concept towards multimodal anticancer agents values with significant standard errors which may be a sign of sec-
[1,30,129–132]. The mode of action of such multimodal anticancer ondary factors such as aqueous solubility and ligand exchange
agents will be influenced by the stability of the complexes formed reactions occurring during the analysis. Furthermore, some modest
which can be exploited to achieve a targeted anticancer effect. antimicrobial activity was observed for gram-positive bacteria.
While in the case of PtIV compounds, reduction of the Pt center Mishra and Singh investigated structural analogues of 41d and
may induce the release of bioactive ligands [133], in other cases 42d for their antibacterial activity [113,138]. However, the lack
the complex may remain intact and exhibit biological activity in of comparison with standard compounds makes it difficult to judge
its synthesized form, for example, by interacting at the interface the potential of the compound type for further investigations.
of two biomolecular targets [132]. On the other hand, slow ligand Introduction of hesperidin led to the preparation of cis-[Ru
exchange could allow for the ligand to exert its function while the (phen)2(hesperidin)](PF6) which, however, was found to be non-
metal fragment may coordinate to another biomolecule. Such fea- cytotoxic, probably due to the high hydrophilicity of the complex
tures can be influenced by the nature of the selected metal center [139]. It was found to inhibit acetylcholinesterase although at rel-
and its oxidation state and allow, in combination with the selection atively high concentrations (IC50 63.6 lM).
of proper ancillary ligands, fine-tuning of the biological activity of Gasser et al. expanded the series of compounds to the 4,7-
the complex. diphenyl-1,10-phenanthroline (DIP) derivatives 43b, 43c, 43e,
and 43f with the corresponding ligands (Fig. 6) [140]. In cytotoxi-
city assays in a broad panel of cell lines, 43e and 43f were signifi-
3.1. Ruthenium compounds of O-donor flavonoids
cantly more active than their analogous free ligands, while 43c was
slightly more cytotoxic than its chrysin ligand c, and 43b and
3.1.1. Ruthenium coordination compounds – From mono- to dinuclear
morin 21 were inactive (Table 1). Comparison of the cytotoxic
anticancer agents
activity of 43f showed that the compound was similarly potent
Ru–bipyridine complexes and their structural analogues have
as the standard anticancer agents cisplatin and doxorubicin. Fur-
been investigated for their DNA binding ability and photochemical
ther comprehensive studies on the cellular uptake revealed that
properties [134]. Unlike cisplatin, their DNA interactions are, how-
the dichlorido precursor [RuII(DIP)2Cl2] was more effectively accu-
ever, based on intercalation rather than covalent bond formation
mulating in MDA-MB-435S cells than 43f but was an order of mag-
and therefore the compounds were expected to feature modes of
nitude less potent in the cell line, which may be influenced by
action different to cisplatin and therewith other tumor types may
several factors, such as the charge state of the complex, labile
be targeted.
ligands, and impact of the genistein 5 ligand on the mode of action.
In order to introduce flavonoids as ligands into polypyridine-Ru
To determine the mechanism of drug uptake (Fig. 7), MDA-MB-
complexes, precursors such as [Ru(bpy)2Cl2] (bpy = 2,20 -bipyridine)
435S and MCF-7 cells were incubated with 43f and for comparison
and [Ru(phen)2X2] (phen = 1,10-phenanthroline; X2 = Cl2, CO3) are
with the precursor [RuII(DIP)2Cl2] at 4 °C, which slows passive dif-
frequently used [135,136]. For example, heteroleptic ruthenium
fusion and active transport as compared to studies at 37 °C, after
bis-bpy 41a-41d and bis-phen 42a-42d (Fig. 6) complexes of quer-
addition of the metabolic inhibitors 2-deoxy-D-glucose and oligo-
cetin 1, morin 21, chrysin 23, and 3-hydroxyflavone 14 were
mycin to block the cellular metabolism by inhibiting ATP produc-
obtained after activation of the ligands with NaOMe (Fig. 6)
tion, of chloroquine and ammonium chloride to slow endocytic
[137]. These complexes featured flavonoids coordinating to the
pathways, and of tetraethylammonium chloride to interfere with
metal center through their carbonyl groups and a neighboring
cation transporters [140]. The studies indicate a passive transport
hydroxyl group after deprotonation to yield 5- or 6-membered
mechanism into the cell for 43f in both cell lines, while the precur-
chelates. The intrinsic fluorescence of tryptophan in BSA was used
sor was actively transported into MDA-MB-435S cells and pas-
to investigate the binding to BSA which was suggested to occur
sively into MCF-7. The mode of action of 43f was suggested to be
through hydrophobic interactions, as would be expected for com-
related to the inhibition of mitochondrial function, glycolysis and
plexes of solely bidentate ligands. Similarly, the ethidium bromide
cytosolic ATP production. The latter feature appears to be specific
substitution assay suggested intercalative binding to DNA. The
to the flavonoid complexes, as structurally related compounds only
cytotoxic activity was assayed in SW620 (metastatic colorectal
inhibited mitochondrial activity but not glycolysis [141–143].
adenocarcinoma), HepG2 (hepatocellular carcinoma), MCF-7
Srishailam et al. employed a phenanthroline linker to connect
(breast adenocarcinoma) and HeLa (cervical carcinoma) cells(Sup-
the bioactive chromone ligand g to bipyridine or phenanthroline
porting Information). Only [Ru(bpy)2(d)]+ 41d was consistently
ruthenium scaffolds [144]. These compounds showed moderate
active across the cell lines investigated and d was the most potent
antiproliferative and antibacterial activities (Supporting
ligand. Interestingly, the authors observed very inconsistent IC50

Table 1
IC50 values (mM) for the free flavonoid ligands, their RuII(4,7-diphenyl-1,10-phenanthroline)(flavonoid) complexes, cisplatin and doxorubicin in the human cell lines MCF-7
(ductal carcinoma), FaDU (pharynx carcinoma), MDA-MB-435S (melanoma), U87 (glioblastoma), RPE-1 (normal retinal pigmented epithelium) HEK293 (embryonic kidney) with
48 h incubation times [140].

Compound IC50 / lM
MCF-7 FaDU MDA-MB-435S U87 RPE-1 HEK293
b >100 >100 >100 >100 >100 >100
c 62.59 ± 3.23 95.06 ± 11.55 79.37 ± 8.13 91.14 ± 13.76 >100 26.80 ± 2.79
e >100 >100 >100 >100 >100 >100
f >100 >100 >100 >100 >100 75.85 ± 0.84
cisplatin 19.69 ± 1.63 5.17 ± 0.21 17.62 ± 0.54 6.97 ± 0.46 39.9 ± 9.14 2.27 ± 0.67
doxorubicin 9.39 ± 1.37 1.55 ± 0.18 5.55 ± 1.37 0.59 ± 0.03 14.9 ± 1.31 0.21 ± 0.03
[Ru(DIP)2Cl2] >50 >50 27.73 ± 5.33 25.59 ± 0.29 3.13 ± 0.28 12.11 ± 1.30
43b >50 >50 >50 >50 >50 >50
43c >50 >50 27.73 ± 5.33 25.59 ± 0.29 23.21 ± 8.08 33.02 ± 3.25
43e >50 38.21 ± 5.22 24.48 ± 1.92 30.72 ± 1.48 19.72 ± 8.23 26.46 ± 3.20
43f 16.67 ± 3.93 5.21 ± 0.73 2.64 ± 0.43 5.21 ± 1.74 2.36 ± 0.77 0.72 ± 0.10

8
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 8. Chemical and molecular structures of the RuII complexes 45–47 and a
dimetallic analogue 48.

zole)]} as an antimetastatic agent. In the preparation of complexes


of chalcone and flavone, Mishra et al. employed cis-[RuCl2(DMSO)4]
Fig. 7. Cellular uptake of (A) [RuII(DIP)2Cl2] and (B) 43f in MDA-MB-435S and MCF- as a Ru precursor which was converted into 45 and the chalcone
7 cells investigated by ICP-MS under different conditions. Reproduced with derivative (Fig. 8) [119]. Analogous Ru(II)-kaempferol (46; Fig. 8)
permission from ref. [140] Ó ACS 2020.
showed potent antiproliferative activity against A549 cells possibly
caused by DNA damage at a concentration of 20 lM perhaps due to
ROS production [146]. Minimal cytotoxic effects were observed in
Information). Additionally, they intercalated into ct-DNA, while WI-38 and HDFa cells for Ru(II)-quercetin (47; Fig. 8) [147]. The di-
not causing photocleavage of plasmid DNA [121]. Similarly, Ru compound 48 (Fig. 8) is structurally related to 47. The molecu-
Gramni et al. functionalized coumarin with a 2,20 -dipicolylamine lar structure of 48 (Fig. 8) indicates a high level of planarity for the
linker to synthesize a fac-trichlorido RuIII complex which can bind flavone ligand which may be beneficial to act as an intercalator.
to ct-DNA but does not have significant DNA cleavage ability or Both derivatives 45 and 48 showed incubation time-dependent
cytotoxicity [145]. cytotoxicity in Dalton lymphoma cells, which seemed to be largely
During the development of RuIII anticancer agents, it became due to the potency of the bidentate ligand [119]. In a similar
apparent that the introduction of DMSO ligands had a beneficial approach, the same group expanded the series of compounds
effect in terms of the biological activity. This resulted in the clinical toward dimetallic 48 with both Ru centers coordinated to one chlo-
development of NAMI-A {imidazolium trans-[RuCl4(DMSO)(imida- rido and three DMSO ligands, either S- or O-bound [148], as well as

9
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

to bisflavone-Ru complexes and flavone complexes with varying


ancillary ligands [149]. While for the former the cytotoxicity was
not investigated, the authors suggest different DNA binding ability
of chalcone and flavone derivatives, although the suggested differ-
ence in conjugation may not be the reason for the contrasting
behavior [148]. For the bisflavone-Ru complexes, issues around
solubility in cell culture medium do not allow to draw final conclu-
sions on the antiproliferative and anti-HIV activity of the com-
pounds while the structural characterization of the complexes
would benefit from further investigations [149]. This observation
demonstrates the often poor solubility observed for Ru(flavone)
complexes which needs to be addressed to make them viable can-
didates for drug development. Roy et al. reported a diverse set of
interesting biological studies for Ru complexes of chrysin and bai-
calein including in vivo data [150,151]. However, it is doubtful that
the complexes were obtained in pure form, given that there were
either no shifts observed in the 1H NMR and IR spectra, the assign-
ments of the peaks in the mass spectra were significantly off com-
pared to the theoretical values, and/or elemental analysis data was
not included. Therefore, all the biological investigations must be
considered under this light and are most probably not meaningful.
For a related class of [bis(3-hydroxyflavone)Ru(DMSO)2] com-
plexes, the cytotoxicity of the flavonol ligands was slightly lower
Fig. 10. Structures of 7,30 ,40 -trihydroxyflavone 53h (top) and chrysin 53c and
than of the respective Ru complexes in the breast cancer cell line tectochrysin 53cMe (bottom) complexes of the trithiacyclononane RuII dimethyl
MCF-7, with the potency of the ligand determining that of the com- sulfoxide scaffold.
plex (Supporting Information) [152]. The effect of metal coordina-
tion was reflected in cell cycle distribution studies where it shifted
a slightly higher number of cells into the G1 phase and more cells which is not surprising given the significant structural differences.
were apoptotic, while the G2 and S phase distributions seemingly Further biological experiments to elucidate the reason for the
declined. However, it remains unclear if the changes were statisti- higher potency in cisplatin-resistant cells revealed a lower level
cally significant. As for the Ru(chrysin)2 complexes mentioned of active caspase-8 in the cisplatin-resistant variant over the wild
above [150], the characterization of the complexes is inconclusive. type cells after treatment with either Ru compound although the
In addition to bisflavonoid complexes with the ligand featuring effect was more pronounced for the methoxy 50OMe over the
an O,O-bidentate chelation motif, Ru compounds with N,O- ethoxy 50OEt derivative [155]. The compounds induced DNA strand
coordinating ligands have been reported [153,154]. Ochocki and breaks in EJ cells and to a lower extent in the resistant variant, and
colleagues reported the dehydrogenative alcoholysis of 3- it is speculated that the oxidative stress induced in both cell types
aminoflavone 49 in methanol or ethanol and the presence of RuCl3 also contributed to the induction of apoptosis. In the preparation of
to yield the [Ru(3-imino-2-alkoxyflavanone)2Cl2] complexes cis-di an analogous complex from RuCl3 and 3-hydroxyiminoflavanone,
chloridobis(3-imino-2-methoxyflavanone)Ru(II)3H2O 50OMe and surprisingly the mixed ligand flavanone/flavone RuII coordination
cis-dichloridobis(3-imino-2-ethoxyflavanone)Ru(II) 2H2O 50OEt. compound 51 was obtained (Fig. 9) [154]. Compound 51 was com-
Independent of the nature of the alkoxy group, the compounds pared to its analogues 50OMe and 50OEt in terms of cytotoxicity and
were less potent than cisplatin in bladder carcinoma EJ cells and was found to be less potent in all cancer cell lines but also in lym-
mouse leukemia cells L1210 but were more active in the phocytes, while the compounds were to a minor degree hemolytic
cisplatin-resistant variants with IC50 values in the low lM range. at higher concentrations. Interestingly, an in silico structure–activ-
Lymphocytes were used as models for healthy cells in which the ity evaluation was investigated in context of Lipinski’s rule of 5,
Ru compounds were an order of magnitude less cytotoxic than cis- where none of the compounds violated more than one drug-
platin. These results suggest a different mode of action to cisplatin likeness rule. The difference in activity was explained by the
reduced lipophilicity of 51 as compared to compounds 50 which
possibly limits the cell membrane determination.
Braga and co-workers prepared 1,4,7-trithiacyclononane ([9]
aneS3) analogues 53 of 45 in which the three DMSO ligands were
substituted with the heterocyclic S-donor ligand and as flavonoids
chrysin c, tectochrysin cMe, and 7,30 ,40 -trihydroxyflavone h were
used [156] (Fig. 10). These structures allow studying the impact
of modification with Ru on the A and B rings. Unexpectedly, com-
plexation to the Ru([9]aneS3) fragment resulted in low aqueous
solubility for 53h which limited its evaluation for cytotoxic activ-
ity. However, the flavonoids used were in all cell lines more potent
than their Ru complexes and in case of chrysin and its Ru complex
51c by an order of magnitude (Supporting Information).
Overall a diverse range of Ru coordination compounds featuring
flavonoid ligands has been reported so far. It appears that in most
cases, if not all, the ligand is the dominating factor when it comes
to their antiproliferative activity. In some cases, metal coordination
Fig. 9. Molecular structure of the mixed ligand 3-hydroxyiminoflavanone/3- seems to have an impact on biological processes, however, for def-
nitrosoflavone RuII coordination compound 51. inite trends clearly more systematic studies are required.

10
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 11. Chemical structures of [Ru(cym)(3-hydroxyflavone)Cl] complexes (54–61) with varying substitution of the flavone B ring, halido leaving groups, arenes and/or
quinolinone.

Table 2
Estimated 50% topoisomerase IIa inhibitory activity (lM) and in vitro anticancer activity (IC50 values in mM) of Ru(cym)Cl complexes 54–60 of 3-hydroxyflavones d in terato-
(CH1/PA-1), colon (SW480), non-small cell lung (A549), urothelial bladder (5637), large cell lung carcinoma (LCLC-103H) and pancreatic adenocarcinoma (DAN-G) compared to
cisplatin.

topoisomerase inhibition IC50 / mM Ref


CH1/PA-1 SW480 A549 5637 LCLC-103H DAN-G
d > 40 mM 1.9 ± 0.2 11 ± 3 25 ± 10 nd nd nd [157]
54d 20–40 mM 2.1 ± 0.2 9.6 ± 1.5 20 ± 2 11 ± 5 13 ± 6 12 ± 2 [157,158]
55d nd 2.8 ± 0.4 12 ± 1 27 ± 4 nd nd nd [164]
56d nd 1.6 ± 0.2 9.6 ± 1.5 16 ± 1 nd nd nd [164]
57d nd 3.2 ± 0.1 12 ± 3 19 ± 1 nd nd nd [164]
58d nd 5.5 ± 1.2 9.2 ± 1.9 28 ± 5 nd nd nd [164]
dp-Me > 40 mM 1.1 ± 0.1 6.3 ± 1.1 81 ± 9 nd nd nd [157]
54dp-Me 20–40 mM 1.8 ± 0.2 7.2 ± 0.5 17 ± 2 5.7 ± 3.2 5.2 ± 0.8 6.6 ± 2.5 [157,158]
dp-F > 40 mM 1.56 ± 0.04 7.0 ± 0.9 37 ± 10 nd nd nd [157]
54dp-F 20–40 mM 1.7 ± 0.4 7.9 ± 2.1 18 ± 1 33 ± 5 5.5 ± 5.2 12 ± 2 [157,158]
dp-Cl 20–40 mM 0.60 ± 0.10 3.7 ± 0.4 7.9 ± 1.2 nd nd nd [157]
54dp-Cl < 20 mM 0.86 ± 0.06 3.8 ± 0.5 9.5 ± 0.5 3.3 ± 1.1 13 ± 1 19 ± 7 [157,158]
55dp-Cl nd 0.86 ± 0.04 3.4 ± 0.4 7.9 ± 0.6 nd nd nd [164]
56dp-Cl nd 1.2 ± 0.3 4.7 ± 0.9 8.9 ± 0.8 nd nd nd [164]
57dp-Cl nd 0.88 ± 0.17 4.7 ± 0.6 7.8 ± 2.5 nd nd nd [164]
58dp-Cl nd 6.3 ± 1.1 21 ± 4 59 ± 1 nd nd nd [164]
54dm-F nd 1.5 ± 0.1 7.0 ± 1.0 15 ± 1 4.3 ± 2.5 4.3 ± 1.1 5.3 ± 1.6 [158]
54do-F nd 4.0 ± 0.8 24 ± 3 30 ± 1 nd nd nd [158]
cisplatin nd 0.14 ± 0.03 3.3 ± 0.4 1.3 ± 0.4 nd nd nd [157]
54dm-Cl nd 1.0 ± 0.1 7.0 ± 0.7 12 ± 2 30 ± 2 5.0 ± 3.5 19 ± 5 [158]
54do-Cl nd 7.9 ± 0.6 26 ± 1 51 ± 5 nd nd nd [158]
54dp-Br nd 1.2 ± 0.2 3.4 ± 0.1 8.6 ± 0.7 12 ± 1 19 ± 6 5.7 ± 1.9 [158]
54dm-Br nd 2.3 ± 0.7 7.2 ± 0.4 17 ± 3 5.9 ± 1.2 16 ± 4 20 ± 5 [158]
dm,p-OMe nd 2.1 ± 0.2 >25 >25 nd nd nd [163]
54dm,p-OMe nd 2.2 ± 0.5 8.7 ± 0.8 18 ± 2 nd nd nd [163]
dm,m-OMe nd 1.4 ± 0.2 >25 >25 nd nd nd [163]
54dm,m-OMe nd 1.5 ± 0.2 4.5 ± 0.2 9.0 ± 0.5 nd nd nd [163]
dm,p,m-OMe a nd 2.0 ± 0.2 8.6 ± 1.5 >25 nd nd nd [163]
54dm,p,m-OMe a nd 2.5 ± 0.3 9.7 ± 1.9 23 ± 5 nd nd nd [163]
do,o-F nd 18 ± 1 >25 >25 nd nd nd [163]
54do,o-F nd 5.1 ± 0.8 20 ± 4 55 ± 15 nd nd nd [163]
dp-CF3 nd 1.0 ± 0.1 2.6 ± 0.2 6.5 ± 1.5 nd nd nd [163]
54dp-CF3 nd nd a nd a nd a nd nd nd [163]
dacetamide nd 7.2 ± 0.3 >25 >25 nd nd nd [163]
54dacetamide nd 5.3 ± 0.5 18 ± 1 21 ± 1 nd nd nd [163]
59 nd 4.0 ± 0.2 14 ± 1 17 ± 2 nd nd nd [164]
60 nd 5.3 ± 0.2 12 ± 2 19 ± 1 nd nd nd [164]
cisplatin nd 0.14 ± 0.03 3.3 ± 0.4 1.3 ± 0.4 nd nd nd
nd, not determined; nd a, insufficient solubility; a
precipitation occurred over 24 h.

11
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

3.1.2. Bioorganoruthenium chemistry: Ru(g6-arene) complexes of O- Efforts to increase the structural diversity of compounds
donor flavonoids resulted in the preparation of analogues with bromido 55 and
The development of RuII(g6-arene) complexes as anticancer iodido 56 leaving groups as well as with toluene 57 and biphenyl
agents has gained a lot of traction over the last decades since 58 as arenes and the quinolinone derivatives 60 and 61 (Fig. 11)
Sadler and Dyson reported their ethylenediamine and pta com- [164]. Introduction of a quinolinone or substitution of the chlorido
plexes, respectively, with significantly differing biological activity ligand for bromido or iodido did not significantly impact the bio-
[29,126]. In an attempt to harvest synergistic activity by combining logical activity of the complexes, again supporting the notion that
bioactive metal centers with ligands with demonstrated biological the antiproliferative activity is dictated by the flavone ligand.
activity, a series of Ru(cym)Cl (cym = g6–p-cymene) complexes of When cym was replaced with a biphenyl ligand as the arene, how-
3-hydroxyflavones (Fig. 11) was synthesized aiming to target the ever, the cytotoxicity dropped significantly for flavonols d and dp-Cl
DNA–topoisomerase complex while maintaining the ability of the (Table 2). This is surprising as often the introduction of larger, con-
complexes to undergo ligand exchange and coordinate to DNA jugated aromatic ligands has led to improved anticancer activity
[157,158], as was reported for other Ru(cym) compounds for Ru(arene) complexes [159].
[126,159]. The Ru complexes were found to indeed inhibit topoiso- The impact of the arene ligand was evaluated in a comparative
merase IIa more effectively than the flavonol compounds [157], study of cym complexes 52 and their N-acetyl-L-phenylalanine
while the cytotoxic activity was often very similar to that of the ethyl ester (AcPheOEt) analogues 61 (Fig. 11) [165]. While com-
bioactive ligand (Table 2), as observed already before for Ru coor- pounds 61 were stable in aqueous solution, they underwent
dination compounds. The para-substituted compounds were the decomposition in DMSO and in presence of amino acids, whereas
most potent cytotoxins in a range of cell lines, in particular 54dp- they formed adducts with 9-ethylguanine as a DNA model through
F and 54dp-Cl, the latter being also the most potent topoisomerase chlorido ligand exchange. Furthermore, it was established that the
IIa inhibitor of the series investigated. Furthermore, the com- compounds damage DNA similarly to cisplatin although to a lesser
pounds were shown to form quickly adducts with 50 -guanosine extent (Fig. 12). While the aqueous solubility of the complexes was
monophosphate, supporting the notion of having designed a mul- significantly higher than found for the cym derivatives, the cyto-
timodal anticancer agent in which a DNA targeting metal moiety toxicity of the compounds was similar or even higher which is
was combined with a protein targeting ligand structure. However, unusual as aqueous solubility often correlates with lipophilicity
it became also apparent that aqueous stability had to be carefully which in turn has significant impact on the cellular uptake. The
assessed as the formation of [(cym)Ru(OH)3Ru(cym)]+ complexes accumulation of 61dp-F in HCT116 cells was significantly higher
was observed by 1H NMR spectroscopy and electrospray than of its cym analogue 54dp-F. This suggests a contribution of
ionization-mass spectrometry over longer incubation periods. Such the Phe-derived ligand beyond that of conventional arene ligands
species are known to be very stable and to form thermodynamic used in the design of piano-stool organometallics.
sinks unreactive towards nucleophiles [160,161]. Compound 54dp-Cl was loaded onto a poly(organo)phosphazene
Analogous structures of Os(cym) and Rh(Cp*) (Cp* = g5- resulting a dendritic structure with three of the metal complexes
pentamethylcyclopentadienyl) have been reported for d, dp-Me, attached to terminal amino groups by substituting the chlorido
dp-F and dp-F [162]. In general, very similar cytotoxic potency ligands in 54dp-Cl [166]. However, the conjugates obtained with
was found as for the Ru(cym) complexes and in fact the flavonol non-flavonol Ru(cym) and Rh(Cp*) compounds showed higher
ligands with, in some cases, the complexes being more active. metal loading in the polymer solution and higher cellular accumu-
The complexes were more reactive towards the amino acid lation in SW480 and CT-26 colon carcinoma cell lines. Further-
L-histidine than towards L-cysteine and glycine. While they did more, the Ru(cym)(flavonol) derivative was not cytotoxic
not bind in protein reactivity studies to cytochrome c, adducts with (Supporting Information) while all the conjugates showed reduced
ubiquitin were detected, in particular for the Rh(Cp*) derivatives. anticancer activity.
Similarly, the harder metal center RhIII preferred in DNA model The Ru(cym) complexes 54d, 54dp-Cl, 54dp-F and the
compound reactivity studies ATP as a binding partner over GTP structurally-related acetonitrile derivative 62d were investigated
which gave a larger number of adducts with the softer RuII and OsII for their photochemical reactivity which resulted in the solvent-
Lewis acids. dependent formation of photoproducts upon irradiation at 300
As flavonoids were reported to inhibit cyclin-dependent kinases and 419 nm (Scheme 1) [167]. For example, ESI-MS showed substi-
(CDKs), the inhibitory potential of the Ru complexes was also tution of the cym ligand with solvent molecules and release of CO.
investigated but no link to cytotoxic activity could be established, The extent of CO release was dependent on the wavelength of irra-
making them unlikely targets [157,158]. Only minor impact on the diation and at 300 nm 0.7 equiv. of CO were detected by GC (0.4
cell cycle distribution of cells was found with a slight G2/M arrest equiv. at 419 nm). Notably, the quantum yields of the complexes
accompanied by a decrease of cells in G0/G1 phase. Exploiting the are very low suggesting that they are normally stable and only dis-
intrinsic fluorescence of 54dp-F, it was found to accumulate in integrate upon irradiation at specific wavelengths. The reactions
the endoplasmic reticulum using confocal fluorescence observed were similar to those reported for the bpy complex 39d
microscopy. under similar conditions [168,169]. In investigations on the anti-
The library of Ru(cym)Cl complexes was expanded with ligands cancer activity of 54d and 62d with and without irradiation it
that carried more than one substituent on the B ring of the flavonol was shown that the photoproducts were non-cytotoxic in Jurkat
(Fig. 11) [163]. In some cases, limited aqueous solubility was and A549 cells. L-Histidine binding studies were performed for
observed for the ligands which was significantly increased by 62d suggesting that the flavone ligand is displaced by L-histidine
metal coordination, making such bioactive compounds more and this process was accompanied by the formation of a
accessible. In molecular structure investigations of a range of com- precipitate.
pounds, it was suggested that a higher degree of planarity of the In addition to investigating the anticancer properties of
flavonol ligand results in higher cytotoxic activity, while lipophilic- organoruthenium(flavonoid) complexes, their inhibitory effects in
ity and electronic effects took supporting roles. The compounds various platelet functions were investigated by Ravishankar et al.
investigated showed antiproliferative activity at slightly higher [110]. Chrysin and other dietary flavonoids are known to exhibit
IC50 values than the more potent derivatives 54dp-Cl and 54dp-F such functions and when combined with a metal center, both the
(Table 2). chrysin and thiochrysin complexes 54c and 54cS (Figs. 11 and

12
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 12. DNA damaging ability in HCT116 cells as determined by flow cytometry after treatment with 54dp-F, 61dp-F, and cisplatin (30 mM each) for 6 h. Reproduced with
permission from ref. [165]. Ó ACS 2018.

Scheme 1. The formation of the main photoinduced reaction products from Ru(cym)(flavone) complexes in acetonitrile.

13), respectively, were shown to have enhanced inhibitory effects than 54c (Fig. 14). These properties were suggested to be attribu-
in platelets under physiological conditions as compared to the ted to different physicochemical properties such as cell permeabil-
ligands, while they exerted slightly reduced effects in washed pla- ity and reactivity to plasma proteins, while they retain the same
telets. This may be explained by higher stability of 54cs, as was also target as found for chrysin itself at low toxicity.
seen for structurally-related, maltol-derived complexes were sub- Yadav and Singh [172] introduced the heteroatoms S and Se
stitution of the carbonyl group resulted in the formation of thioma- into the chrysin c scaffold by factionalizing it in 7-position with
ltolato Ru compounds which were more potent anticancer agents thio and seleno ethers (63a–63d, Fig. 13) for a synergistic effect
due to higher stability under aqueous conditions and in presence with regard to radical scavenging and other biological properties
of biological ligands [161,170,171]. The thiochrysin complex 54cS [173]. DNA and protein binding were demonstrated using spectro-
reduced thrombus growth and volume to a 10–25% higher degree scopic methods, suggesting electrostatic interactions with DNA

13
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Fig. 13. Structures of RuII(cym) seleno- and thioether chrysin complexes.

and with HSA by hydrogen bonding and van der Waals forces,
while docking was employed to elucidate preferred binding sites.
The Se derivatives were found to have higher binding constants Fig. 15. The structures of ruthenium(II) complexes with aminoflavones.
to DNA than their sulfur analogues. The potential of the com-
pounds as anticancer agents is difficult to assess as the results from
a sulforhodamine B cytotoxicity assay were not reported as IC50 reaction of 3-aminoflavone 49 derivatives with ruthenium(III)
values. compounds [153]. 7-Aminoflavone 64 and 6-aminoflavone 65
Overall, organoruthenium compounds have shown a lot of formed complexes after reaction with the [Ru(cym)Cl2]2 dimeric
potential for anticancer chemotherapeutics development and precursor and their in vitro properties were studied in melanoma
incorporation of a flavonoid ligand has in some cases resulted in cell lines (Fig. 15) [175]. Compound 65 and the ruthenium deriva-
synergistic biological activity. However, there are in some cases tive 69 had moderate activity against melanoma cells like A375
questions around solubility and stability in biological media which (human melanoma cells with high metastatic potential), DMBC11
need to be addressed. The substitution of coordinating carbonyl (Superficial Spreading Melanoma (SSM)), and DMBC12 (Nodular
groups for thiocarbonyls may be a way of addressing the latter, Melanoma (NM)), with poor activity against leukemia K562 cells.
while the rich decoration of the flavonoid scaffold with hydroxyl Pastuszko et al. [174,176] also investigated the cytotoxicity of
moieties leaves enough of flexibility for the synthetic chemist to the Ru(arene) complexes of 6-aminoflavone 65 and
improve aqueous solubility. In general, the use of comparable 7-aminoflavone 64 with cym, benzene and hexamethylbenzene
methods would clearly enhance the development prospect of the arene derivatives and chlorido and iodido ligands as leaving groups
compound type and further studies have to focus on obtaining in 66–71. The highest cytotoxic activities were observed for ruthe-
in vivo efficacy data to be able to confidently judge the potential nium(II) complexes with ligand 64 with the IC50 values being sim-
of the compound class for further development. ilar for both melanoma cell lines (DMBC11 and DMBC12) with
DMBC12 cells being more sensitive than DMBC11. Recently, Mucha
3.2. Ru(II)/(III) complexes of aminoflavonoids and coworkers [177] investigated the cytotoxic effect of ruthenium
(II) and copper(II) complexes with aminoflavone ligands in the
In contrast to hydroxyflavones, aminoflavones do not occur nat- human cancer cell lines WM 115 (melanoma cell line), HL-60
urally. Pastuszko et al. [174] synthesized RuII(arene) complexes of (promyelocytic leukemia cells), NALM-6 (peripheral blood
aminoflavone derivatives while Ochocki et al. investigated the leukemia cells) and COLO 205 (colon adenocarcinoma) (Table 3).

Fig. 14. Comparison of the time-dependent effects of chrysin c and the thiochrysin Ru complex 54cs (both at 100 lM concentrations) in the thrombus formation under
arterial flow conditions as recorded by fluorescence microscopy. Reproduced with permission from ref. [110]. Ó Nature 2017.

14
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

Table 3
The in vitro anticancer activity (IC50 values in mM) of Ru(cym)Cl complexes 66–71 and the respective free ligands 64 and 65 in melanoma (DMBC11, DMBC12, and WM-115),
promyelocytic leukemia (HL-60), peripheral blood leukemia (NALM-6), and colon (COLO 205) cancer cell lines (DMCB11 & DMCB12 from [174,176] and HL-60 NALM-6, WM-115
& COLO 205 from [177].

Compound IC50 / lM
DMBC11 DMBC12 HL-60 NALM-6 WM-115 COLO 205
64 nd nd 54.5 ± 2.0 54.5 ± 2.2 90.6 ± 5.4 4.73 ± 4.6
65 nd nd 272.4 ± 38.2 66.5 ± 5.3 699.1 ± 53.6 316.9 ± 30.4
66 nd nd 79.0 ± 4.0 65.2 ± 1.3 412.1 ± 49.7 175.6 ± 29.0
66a 13.04 1.13 nd nd nd - nd
67 4.04 2.53 140.7 ± 11.4 75.8 ± 3.7 474.2 ± 40.8 381.5 ± 44.4
68 13.04 2.31 91.2 ± 5.3 62.8 ± 2.9 334.5 ± 40.8 353.0 ± 37.3
69 19.15 0.96 276.8 ± 37.2 67.0 ± 4.7 394.2 ± 26.2 406.4 ± 39.1
70 nd nd nd nd nd nd
71 nd nd nd nd nd nd

The Ru(II) complexes were less cytotoxic than the respective cop- Declaration of Competing Interest
per(II) compounds (Supporting Information), with the copper(II)
species being more potent than the free ligands. In many cases, The authors declare that they have no known competing finan-
the cytotoxicity of the Ru(II) complexes are lower than their corre- cial interests or personal relationships that could have appeared
sponding ligands against the NALM-6 cell line being the most to influence the work reported in this paper.
sensitive.
Acknowledgements
4. Conclusions
Financial support from Medical University of Lodz (grant No.
In this review, the structural and biological features of Ru(II)/ 503/3-066-02/503-31-001 to E. Budzisz), University of Lodz (grant
(III) complexes with flavonoid ligands were discussed. Flavonoids No. B1811100000050.01 to M. Malecka) and the University of
are commonly found in plants and possess medicinal and phar- Auckland is acknowledged.
maceutical properties. Their therapeutic targets have been sug-
gested as kinases, apoptotic proteins, growth factors, and
matrix metalloproteinases. Molecular docking investigations can Appendix A. Supplementary data
be helpful by predicting conformational changes of the protein
following ligand binding, thereby guiding efforts towards suc- Supplementary data to this article can be found online at
cessful synthesis of novel anticancer drugs. Due to the promising https://doi.org/10.1016/j.ccr.2021.213849.
biological properties of flavonoids, coordination to metal centers
such as Ru has been suggested to result in synergistic effects. References
Indeed, in some cases, this design principle resulted in com-
pounds with improved antiproliferative and/or enzyme inhibi- [1] M. Grazul, E. Budzisz, Biological activity of metal ions complexes of
chromones, coumarins and flavones, Coord. Chem. Rev. 253 (21-22) (2009)
tory activity than observed for the flavonoids themselves. 2588–2598, https://doi.org/10.1016/j.ccr.2009.06.015.
Importantly, it is not yet possible to deduce unambiguous struc- [2] E. Budzisz, B. Keppler, G. Giester, M. Wozniczka, A. Kufelnicki, B. Nawrot,
ture–activity relationships. In most compound classes, indepen- Synthesis, crystal structure and biological characterization of a novel
palladium(II) complex with a coumarin-derived ligand, Eur. J. Inorg. Chem.
dent of the structure of the metal complex, being 2004 (22) (2004) 4412–4419, https://doi.org/10.1002/ejic.200400483.
organometallic (pseudo)half-sandwich or octahedral coordination [3] J.H. Choi, K.S. Ahn, J. Kim, Y.S. Hong, Enhanced induction of Bax gene
compounds, derivatives have been shown to be potent cytotox- expression in H460 and H1299 cells with the combined treatment of cisplatin
and adenovirus mediated wt-p53 gene transfer, Exp. Mol. Med. 32 (1) (2000)
ins in cancer cells. The coordination mode of the flavonoid ligand
23–28, https://doi.org/10.1038/emm.2000.5.
to the metal center seems to have at least in some cases impact [4] E. Zyner, J. Graczyk, J. Ochocki, Pt(II) and Pd(II) complexes of 3-aminoflavone:
on the biological activity, with ligand exchange reactions nega- in vitro and in vivo evaluation, Pharmazie. 54 (1999) 945–946. http://www.
tively impacting on their biological properties. Hence the biolog- ncbi.nlm.nih.gov/pubmed/10631759.
_
[5] B. Zurowska, A. Erxleben, Ł. Glinka, M. Łe˛czycka, E. Zyner, J. Ochocki,
ical activity of many complexes was driven by the flavonoid Synthesis, spectroscopy and magnetism of novel metal complexes of 3-
ligands themselves potentially following cleavage from the metal aminoflavone (3-af). X-ray crystal structure of 3-af and [Cu(3-af)2(NO3)2],
center. These are important implications when considering such Inorganica Chim. Acta. 362 (3) (2009) 739–744, https://doi.org/10.1016/j.
ica.2008.04.014.
compounds for further investigation as chemotherapeutics, as [6] R.K. Graham, Y.u. Deng, E.J. Slow, B. Haigh, N. Bissada, G.e. Lu, J. Pearson, J.
are other physicochemical properties such as solubility and Shehadeh, L. Bertram, Z. Murphy, S.C. Warby, C.N. Doty, S. Roy, C.L.
lipophilicity, which influence the drug-likeness of the complexes. Wellington, B.R. Leavitt, L.A. Raymond, D.W. Nicholson, M.R. Hayden,
Cleavage at the Caspase-6 Site Is Required for Neuronal Dysfunction and
While there are encouraging signs for some of Ru(flavonoid) Degeneration Due to Mutant Huntingtin, Cell 125 (6) (2006) 1179–1191,
complex types, future success in ruthenium-flavonoid drug https://doi.org/10.1016/j.cell.2006.04.026.
design obviously depends heavily on the choice of the ligands, [7] K.S. Ghosh, T.K. Maiti, A. Mandal, S. Dasgupta, Copper complexes of (-)-
epicatechin gallate and (-)-epigallocatechin gallate act as inhibitors of
defining the reactivity of the compounds, stabilization of the oxi- Ribonuclease A, FEBS Lett. 580 (2006) 4703–4708, https://doi.org/10.1016/j.
dation state of ruthenium and the overall lipophilicity of the febslet.2006.07.054.
complex and therewith improving drug-like properties. More- [8] J.H. Fox, J.A. Kama, G. Lieberman, R. Chopra, K. Dorsey, V. Chopra, I. Volitakis,
R.A. Cherny, A.I. Bush, S. Hersch, K. Gwinn-Hardy, Mechanisms of copper ion
over, for the compound class, there is a lack of in vivo data avail-
mediated Huntington’s disease progression, PLoS ONE 2 (3) (2007) e334,
able, which would be the next step to assess the viability of the https://doi.org/10.1371/journal.pone.0000334.
approach for further development. Overall, it is apparent that [9] M.A. Ebrahimzadeh, F. Pourmorad, A.R. Bekhradnia, Iron chelating activity,
metal complexes of flavonoids possess interesting biological phenol and flavonoid content of some medicinal plants from Iran, African J.
Biotechnol. 7 (2008) 3188–3192.
properties, which could be significant for their applications in [10] C. Gardi, B. Arezzini, V. Fortino, M. Comporti, Effect of free iron on collagen
medicine and pharmacy. synthesis, cell proliferation and MMP-2 expression in rat hepatic stellate

15
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

cells, Biochem. Pharmacol. 64 (7) (2002) 1139–1145, https://doi.org/10.1016/ Biochem. Pharmacol. 38 (10) (1989) 1617–1624, https://doi.org/10.1016/
S0006-2952(02)01257-1. 0006-2952(89)90309-2.
[11] Y. Zhao, H. Li, Z. Gao, H. Xu, Effects of dietary baicalin supplementation on [37] L.-T. Lee, Y.-T. Huang, J.-J. Hwang, A.-L. Lee, F.-C. Ke, C.-J. Huang, C.
iron overload-induced mouse liver oxidative injury, Eur. J. Pharmacol. 509 (2- Kandaswami, P.-P. Lee, M.-T. Lee, Transinactivation of the epidermal growth
3) (2005) 195–200, https://doi.org/10.1016/j.ejphar.2004.11.060. factor receptor tyrosine kinase and focal adhesion kinase phosphorylation by
[12] A. Nakayama, M. Hiromura, Y. Adachi, H. Sakurai, Molecular mechanism of dietary flavonoids: Effect on invasive potential of human carcinoma cells,
antidiabetic zinc-allixin complexes: Regulations of glucose utilization and Biochem. Pharmacol. 67 (11) (2004) 2103–2114, https://doi.org/10.1016/j.
lipid metabolism, J. Biol. Inorg. Chem. 13 (5) (2008) 675–684, https://doi.org/ bcp.2004.02.023.
10.1007/s00775-008-0352-0. [38] A. Tawani, A. Kumar, Structural Insight into the interaction of Flavonoids with
[13] L.H. Cazarolli, L. Zanatta, A.P. Jorge, E. de Sousa, H. Horst, V.M. Woehl, M.G. Human Telomeric Sequence, Sci. Rep. 5 (2015) 1–13, https://doi.org/10.1038/
Pizzolatti, B. Szpoganicz, F.R.M.B. Silva, Follow-up studies on glycosylated srep17574.
flavonoids and their complexes with vanadium: Their anti-hyperglycemic [39] M.T.L.Y.T. Huang, J.J. Hwang, P.P. Lee, F.C. Ke, J.H. Huang, C.J. Huang, C.
potential role in diabetes, Chem. Biol. Interact. 163 (3) (2006) 177–191, Kandaswami, E. Jr Middleton, Effects of luteolin and quercetin, inhibitors of
https://doi.org/10.1016/j.cbi.2006.07.010. tyrosine kinase, on cell growth and metastasis-associated properties in A431
[14] A.S. Clark, J.M. Fagan, W.E. Mitch, Selectivity of the insulin-like actions of cells overexpressing epidermal growth factor receptor, J. Pharmacol. (1999)
vanadate on glucose and protein metabolism in skeletal muscle, Biochem. J. 999–1010, https://doi.org/10.1074/jbc.275.12.8806.
232 (1985) 273–276, https://doi.org/10.1042/bj2320273. [40] T. Akiyama, J. Ishida, S. Nakagawa, H. Ogawara, S. Watanabe, N. Itoh, M.
[15] K.C. Ong, H.-E. Khoo, Insulinomimetic effects of myricetin on lipogenesis and Shibuya, Y. Fukami, Genistein, a specific inhibitor of tyrosine-specific protein
glucose transport in rat adipocytes but not glucose transporter translocation, kinases, J. Biol. Chem. 262 (12) (1987) 5592–5595, https://doi.org/10.1016/
Biochem. Pharmacol. 51 (4) (1996) 423–429, https://doi.org/10.1016/0006- S0021-9258(18)45614-1.
2952(95)02195-7. [41] G.T. Merlino, Y.H. Xu, S. Ishii, A.J.L. Clark, K. Semba, K. Toyoshima, T.
[16] B. Singh, A. Kumar, A.K. Malik, Flavonoids biosynthesis in plants and its Yamamoto, I. Pastan, Amplification and enhanced expression of the
further analysis by capillary electrophoresis, Electrophoresis 38 (6) (2017) epidermal growth factor receptor gene in A431 human carcinoma cells,
820–832, https://doi.org/10.1002/elps.201600334. Science (80-.). 224 (1984) 415–417. https://doi.org/10.1126/science.6200934.
[17] W. Bors, W. Heller, C. Michel, The Chemistry of Flavonoids. Flavonoids in [42] Z.D. F. Traganos, B. Ardelt, M. Halko, S. Bruno, Effects of genistin of the growth
Health and Disease, in: Rice-Evans (Ed.), Chem. Flavonoids, Marcel Dekker , and cell cycle progression of normal human lumphocytes and human
1998: pp. 111–136. https://doi.org/10.1155/2013/162750. leukemic MOLT-4 and HL-60 cells, 52 (1999) 6200–6208.
[18] P. Pietta, Flavonoids in Medicinal Plants. Flavonoids in Health and Disease, [43] C. Kandaswami, E. Perkins, G. Drzewiecki, D.S. Soloniuk, E. Middleton,
Marcel Dekker, 1998. Differential inhibition of proliferation of human squamous cell carcinoma,
[19] S. Shukla, Higher plant flavonoids: biosynthesis and chemical ecology, in: M. gliosarcoma and embryonic fibroblast-like lung cells in culture by plant
Berhow, S. Vaughn (Eds.), Allelochem. Interact. Dakshini, In Princip, CRC Press flavonoids, Anticancer. Drugs. 3 (5) (1992) 525–530, https://doi.org/10.1097/
New York, 1999: pp. 423–437. 00001813-199210000-00013.
[20] Y.Y. Lin, K.J. Ng, S. Yang, Characterization of flavonoids by liquid [44] C. Kandaswami, E. Perkins, D.S. Soloniuk, G. Drzewiecki, E. Middleton,
chromatography—tandem mass spectrometry, J. Chromatogr. A. 629 (2) Antitproliferative effects of citrus flavonoids on a human squamous cell
(1993) 389–393, https://doi.org/10.1016/0021-9673(93)87054-P. carcinoma in vitro, Cancer Lett. 56 (2) (1991) 147–152, https://doi.org/
[21] S. Kumar, A.K. Pandey, Chemistry and biological activities of flavonoids: An 10.1016/0304-3835(91)90089-Z.
overview, Sci. World J. 2013 (2013) 1–16, https://doi.org/10.1155/2013/ [45] M. Leone, D. Zhai, S. Sareth, S. Kitada, J.C. Reed, M. Pellecchia, Cancer
162750. prevention by tea polyphenols is linked to their direct inhibition of
[22] T.-Y. Wang, Q. Li, K.-S. Bi, Bioactive flavonoids in medicinal plants: Structure, antiapoptotic Bcl-2-family proteins, Cancer Res. (2003) 8118–8121.
activity and biological fate, Asian J. Pharm. Sci. 13 (1) (2018) 12–23, https:// [46] M.A. Avila, J.A. Velasco, J. Cansado, V. Notano, Quercetin Mediates the Down-
doi.org/10.1016/j.ajps.2017.08.004. Regulation of Mutant p53 in the Human Breast Cancer Cell Line MDA-MB468,
[23] M. Abid, F. Shamsi, A. Azam, Ruthenium Complexes: An Emerging Ground to Cancer Res. 54 (1994) 2424–2428.
the Development of Metallopharmaceuticals for Cancer Therapy, Mini- [47] L.R. Chu, D.-C,. JUneja, General chemical composition of green tea and its
Reviews Med. Chem. 16 (2015) 772–786, https://doi.org/10.2174/ infusion, in: Chem. Appl. Green Tea, CRC Press New York, 1997: pp. 13–22.
1389557515666151001142012. https://doi.org/10.11402/cookeryscience.50.182.
[24] S. Thota, Editorial (Thematic Issue: Anticancer Ruthenium Complexes in Drug [48] Sanjeev Shukla, Sanjay Gupta, Molecular Mechanisms for Apigenin-Induced
Discovery and Medicinal Chemistry), Mini-Reviews Med. Chem. 16 (2016) Cell-Cycle Arrest and Apoptosis of Hormone Refractory Human Prostate
771–771. https://doi.org/10.2174/138955751610160503003405. Carcinoma DU145 Cells, Mol. Carcinog. 39 (2) (2004) 114, https://doi.org/
[25] H.M. Southam, J.A. Butler, J.A. Chapman, R.K. Poole, The Microbiology of 10.1002/mc.10168.
Ruthenium Complexes, in: Adv. Microb. Physiol., 2017: pp. 1–96. https://doi. [49] Marc E. Bracke, Vincent Castronovo, Rita M.L. Van Cauwenberge, Peter
org/10.1016/bs.ampbs.2017.03.001. Coopman, Luc Vakaet, Pawel Strojny, Jean-Michel Foidart, Marc M. Mareel,
[26] C.G. Hartinger, P.J. Dyson, Bioorganometallic chemistry—from teaching The anti-invasive flavonoid (+)-catechin binds to laminin and abrogates the
paradigms to medicinal applications, Chem. Soc. Rev. 38 (2) (2009) 391– effect of laminin on cell morphology and adhesion, Exp. Cell Res. 173 (1)
401, https://doi.org/10.1039/B707077M. (1987) 193–205, https://doi.org/10.1016/0014-4827(87)90345-4.
[27] U. Jungwirth, C.R. Kowol, B.K. Keppler, C.G. Hartinger, W. Berger, P. Heffeter, [50] Eleni Bagli, Maria Stefaniotou, Lucia Morbidelli, Marina Ziche, Konstantinos
Anticancer activity of metal complexes: Involvement of redox processes, Psillas, Carol Murphy, Theodore Fotsis, Luteolin inhibits vascular endothelial
Antioxidants Redox Signal. 15 (4) (2011) 1085–1127, https://doi.org/10.1089/ growth factor-induced angiogenesis; inhibition of endothelial cell survival and
ars.2010.3663. proliferation by targeting phosphatidylinositol 30 -kinase activity, Cancer Res.
[28] P.J. Dyson, G. Sava, Metal-based antitumour drugs in the post genomic era, 64 (21) (2004) 7936–7946, https://doi.org/10.1158/0008-5472.CAN-03-3104.
Dalt. Trans. (16) (2006) 1929, https://doi.org/10.1039/b601840h. [51] Graziano Lolli, Giorgio Cozza, Marco Mazzorana, Elena Tibaldi, Luca Cesaro,
[29] S.J. Dougan, P.J. Sadler, The design of organometallic ruthenium arene Arianna Donella-Deana, Flavio Meggio, Andrea Venerando, Cinzia Franchin,
anticancer agents, Chimia (Aarau). 61 (11) (2007) 704–715, https://doi.org/ Stefania Sarno, Roberto Battistutta, Lorenzo A. Pinna, Inhibition of protein
10.2533/chimia.2007.704. kinase CK2 by flavonoids and tyrphostins. a structural insight, Biochemistry
[30] R.G. Kenny, C.J. Marmion, Toward Multi-Targeted Platinum and Ruthenium 51 (31) (2012) 6097–6107, https://doi.org/10.1021/bi300531c.
Drugs - A New Paradigm in Cancer Drug Treatment Regimens?, Chem. Rev. [52] Takeshi Yokoyama, Yuto Kosaka, Mineyuki Mizuguchi, Structural Insight into
119 (2) (2019) 1058–1137, https://doi.org/10.1021/acs.chemrev.8b00271. the Interactions between Death-Associated Protein Kinase 1 and Natural
[31] E. Alessio, Thirty Years of the Drug Candidate NAMI-A and the Myths in the Flavonoids, J. Med. Chem. 58 (18) (2015) 7400–7408, https://doi.org/10.1021/
Field of Ruthenium Anticancer Compounds: A Personal Perspective, Eur. J. acs.jmedchem.5b00893.
Inorg. Chem. 2017 (2017) 1549–1560, https://doi.org/10.1002/ [53] H.-L. Peng, W.-C. Huang, S.-C. Cheng, C.-J. Liou, Fisetin inhibits the generation
ejic.201600986. of inflammatory mediators in interleukin-1b–induced human lung epithelial
[32] C. Kanadaswami, L.T. Lee, P.P.H. Lee, J.J. Hwang, F.C. Ke, Y.T. Huang, M.T. Lee, cells by suppressing the NF-jB and ERK1/2 pathways, Int.
The antitumor activities of flavonoids, In Vivo (Brooklyn). 19 (2005) 895–910. Immunopharmacol. 60 (2018) 202–210, https://doi.org/10.1016/j.
https://doi.org/895-910. 0258-851X/2005. intimp.2018.05.004.
[33] L. Gamet-Payrastre, S. Manenti, M.-P. Gratacap, J. Tulliez, H. Chap, B. [54] Lei Chen, Hui Teng, Zhen Jia, Maurizio Battino, Anca Miron, Zhiling Yu, Hui
Payrastre, Flavonoids and the inhibition of PKC and PI 3-kinase, Gen. Cao, Jianbo Xiao, Intracellular signaling pathways of inflammation modulated
Pharmacol. 32 (3) (1999) 279–286, https://doi.org/10.1016/S0306-3623(98) by dietary flavonoids: The most recent evidence, Crit. Rev. Food Sci. Nutr. 58
00220-1. (17) (2018) 2908–2924, https://doi.org/10.1080/10408398.2017.1345853.
[34] T.-B. Kang, N.-C. Liang, Studies on the inhibitory effects of quercetin on the [55] N. Yahfoufi, N. Alsadi, M. Jambi, C. Matar, The immunomodulatory and anti-
HL-60 leukemia cells, Biochem. Pharmacol. 54 (1997) 1013–1018, https://doi. inflammatory role of polyphenols, Nutrients. 10 (2018) 1618, https://doi.org/
org/10.1016/S0006-2952(97)00260-8. 10.3390/nu10111618.
[35] Y. Graziani, E. Erikson, R.L. Erikson, The effect of quercetin on the [56] Reetesh Kumar, Icaro P Caruso, Anwar Ullah, Marinonio Lopes Cornelio,
phosphorylatio activity of the Rous sarcoma virus transforming gene Marcelo Andres Fossey, Fatima Pereira de Souza, Raghuvir Krishnaswamy
product in vitro and in vivo, Eur. J. Biochem. 135 (3) (1983) 583–589, Arni, Exploring the Binding Mechanism of Flavonoid Quercetin to
https://doi.org/10.1111/j.1432-1033.1983.tb07692.x. Phospholipase A2: Fluorescence Spectroscopy and Computational Approach,
[36] P.C. Ferriola, V. Cody, E. Middleton, Protein kinase C inhibition by plant Eur, J. Exp. Biol. 07 (05) (2017), https://doi.org/10.21767/2248-
flavonoids. Kinetic mechanisms and structure-activity relationships, 921510.21767/2248-9215.100033.

16
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

[57] M. Novo Belchor, H. Hessel Gaeta, C. Fabri Bittencourt Rodrigues, C. Ramos da Curr. Cancer Drug Targets. 6 (2006) 613–622, https://doi.org/10.2174/
Cruz Costa, D. de Oliveira Toyama, L. Domingues Passero, M. Dalastra 156800906778742451.
Laurenti, M. Hikari Toyama, Evaluation of Rhamnetin as an Inhibitor of the [77] J. Jawien, R. Korbut, The current view on the role of leukotrienes in
Pharmacological Effect of Secretory Phospholipase A2, Molecules. 22 (2017) atherogenesis, J. Physiol. Pharmacol. 61 (2010) 647–650.
1441. https://doi.org/10.3390/molecules22091441. [78] Oliver Werz, Dieter Steinhilber, Therapeutic options for 5-lipoxygenase
[58] Zuzana Hanáková, Jan Hošek, Zsófia Kutil, Veronika Temml, Přemysl Landa, inhibitors, Pharmacol. Ther. 112 (3) (2006) 701–718, https://doi.org/
Tomáš Vaněk, Daniela Schuster, Stefano Dall’Acqua, Josef Cvačka, Ondřej 10.1016/j.pharmthera.2006.05.009.
Polanský, Karel Šmejkal, Anti-inflammatory Activity of Natural Geranylated [79] W. Berger, M.T.M. De Chandt, C.B. Cairns, Zileuton: Clinical implications of 5-
Flavonoids: Cyclooxygenase and Lipoxygenase Inhibitory Properties and Lipoxygenase inhibition in severe airway disease, Int. J. Clin. Pract. 61 (2007)
Proteomic Analysis, J. Nat. Prod. 80 (4) (2017) 999–1006, https://doi.org/ 663–676, https://doi.org/10.1111/j.1742-1241.2007.01320.x.
10.1021/acs.jnatprod.6b01011. [80] Swati Singh, Manika Awasthi, Veda P. Pandey, Upendra N. Dwivedi,
[59] D.M. González Mosquera, Y. Hernández Ortega, P.L. Fernández, Y. González, D. Lipoxygenase directed anti-inflammatory and anti-cancerous secondary
Doens, Y. Vander Heyden, K. Foubert, L. Pieters, Flavonoids from Boldoa metabolites: ADMET-based screening, molecular docking and dynamics
purpurascens inhibit proinflammatory cytokines (TNF-a and IL-6) and the simulation, J. Biomol. Struct. Dyn. 35 (3) (2017) 657–668, https://doi.org/
expression of COX-2, Phyther, Res. 32 (9) (2018) 1750–1754, https://doi.org/ 10.1080/07391102.2016.1159985.
10.1002/ptr.6104. [81] O. Rådmark, B. Samuelsson, Microsomal prostaglandin e synthase-1 and 5-
[60] Fei Gao, Dong Wei, Tao Bian, Ping Xie, Jian Zou, Huijun Mu, Bin Zhang, lipoxygenase: Potential drug targets in cancer, J. Intern. Med. 268 (2010) 5–
Xiaoyan Zhou, Genistein attenuated allergic airway inflammation by 14, https://doi.org/10.1111/j.1365-2796.2010.02246.x.
modulating the transcription factors T-bet, GATA-3 and STAT-6 in a murine [82] A. Zahir, A. Jossang, B. Bodo, J. Provost, J.-P. Cosson, T. Sévenet, DNA
model of asthma, Pharmacology 89 (3-4) (2012) 229–236, https://doi.org/ topoisomerase I inhibitors: Cytotoxic flavones from Lethedon tannaensis, J.
10.1159/000337180. Nat. Prod. 59 (1996) 701–703, https://doi.org/10.1021/np960336f.
[61] Changda Liu, David Weir, Paula Busse, Nan Yang, Zhenwen Zhou, Charles [83] Heather L. Howie, Rachel A. Katzenellenbogen, Denise A. Galloway,
Emala, Xiu-Min Li, The flavonoid 7,40 -dihydroxyflavone inhibits MUC5AC Papillomavirus E6 proteins, Virology 384 (2) (2009) 324–334, https://doi.
gene expression, production, and secretion via regulation of NF-jB, STAT6, org/10.1016/j.virol.2008.11.017.
and HDAC2, Phyther. Res. 29 (6) (2015) 925–932, https://doi.org/10.1002/ [84] P. Kulshrestha, V.K. Jatav, S. Sharma, Screening of effective herbal compounds
ptr.5334. for inhibition of human papillomavirus type 16 E6 protein, Imternational Res,
[62] Douglas B. Kitchen, Hélène Decornez, John R. Furr, Jürgen Bajorath, Docking J. Humanit. Eng. Pharm. Sci. 2 (2015).
and scoring in virtual screening for drug discovery: Methods and [85] Naglaa Ashmawy, Mohamed Ashour, Michael Wink, Mohamed El-Shazly,
applications, Nat. Rev. Drug Discov. 3 (11) (2004) 935–949, https://doi.org/ Fang-Rong Chang, Noha Swilam, Ashraf Abdel-Naim, Nahla Ayoub,
10.1038/nrd1549. Polyphenols from Erythrina crista-galli: Structures, molecular docking and
[63] Q.-N. Tran, H.R. Arabnia, Emerging Trends in Computational Biology, phytoestrogenic activity, Molecules 21 (6) (2016) 726, https://doi.org/
Bioinformatics, and Systems Biology: Algorithms and Software Tools 10.3390/molecules21060726.
(2015), https://doi.org/10.1016/C2014-0-02761-X. [86] L. Celik, J.D.D. Lund, B. Schiøtt, Conformational dynamics of the estrogen
[64] Sérgio Filipe Sousa, Pedro Alexandrino Fernandes, Maria João Ramos, Protein- receptor a: Molecular dynamics simulations of the influence of binding site
ligand docking: Current status and future challenges, Proteins Struct. Funct. structure on protein dynamics, Biochemistry 46 (2007) 1743–1758, https://
Genet. 65 (1) (2006) 15–26, https://doi.org/10.1002/prot.21082. doi.org/10.1021/bi061656t.
[65] H. Gohlke, G. Klebe, Approaches to the description and prediction of the [87] Yacong Bo, Jinfeng Sun, Mengmeng Wang, Jizhe Ding, Quanjun Lu, Ling Yuan,
binding affinity of small-molecule ligands to macromolecular receptors, Dietary flavonoid intake and the risk of digestive tract cancers: A systematic
Angew. Chemie - Int. Ed. 41 (2002) 2644–2676, https://doi.org/10.1002/ review and meta-analysis, Sci. Rep. 6 (1) (2016), https://doi.org/10.1038/
1521-3773(20020802)41:15<2644::AID-ANIE2644>3.0.CO;2-O. srep24836.
[66] Yu Shan, Yan Cheng, Yi Zhang, Fu-Qin Guan, Hao Sun, Xing-cong Ren, Yu [88] G.-L. Chen, M.-X. Fan, J.-L. Wu, N. Li, M.-Q. Guo, Antioxidant and anti-
Chen, Xu Feng, Jin-Ming Yang, Triticuside a, a dietary flavonoid, inhibits inflammatory properties of flavonoids from lotus plumule, Food Chem. 277
proliferation of human breast cancer cells via inducing apoptosis, Nutr. (2019) 706–712, https://doi.org/10.1016/j.foodchem.2018.11.040.
Cancer. 65 (6) (2013) 891–899, https://doi.org/10.1080/ [89] Xican Li, Qian Jiang, Tingting Wang, Jingjing Liu, Dongfeng Chen, Comparison
01635581.2013.802001. of the antioxidant effects of quercitrin and isoquercitrin: Understanding the
[67] Muhammad Furqan Akhtar, Ammara Saleem, Azhar Rasul, Mirza Muhammad role of the 600 -OH group, Molecules 21 (9) (2016) 1246, https://doi.org/
Faran Ashraf Baig, May Bin-Jumah, Mohamed M. Abdel Daim, Anticancer 10.3390/molecules21091246.
natural medicines: An overview of cell signaling and other targets of [90] Y.-Q. Guo, G.-H. Tang, L.-L. Lou, W. Li, B. Zhang, B. Liu, S. Yin, Prenylated
anticancer phytochemicals, Eur. J. Pharmacol. 888 (2020) 173488, https:// flavonoids as potent phosphodiesterase-4 inhibitors from Morus alba:
doi.org/10.1016/j.ejphar.2020.173488. Isolation, modification, and structure-activity relationship study, Eur. J.
[68] Danielle G Smith, Tapiwanashe Magwere, Susan A Burchill, Oxidative stress Med. Chem. 144 (2018) 758–766, https://doi.org/10.1016/j.
and therapeutic opportunities: Focus on the Ewing’s sarcoma family of ejmech.2017.12.057.
tumors, Expert Rev. Anticancer Ther. 11 (2) (2011) 229–249, https://doi.org/ [91] B. Wahlang, C. McClain, S. Barve, L. Gobejishvili, Role of cAMP and
10.1586/era.10.224. phosphodiesterase signaling in liver health and disease, Cell. Signal. 49
[69] Hyeryoung Jung, Soon Young Shin, Yearam Jung, Thao Anh Tran, Hye Ok Lee, (2018) 105–115, https://doi.org/10.1016/j.cellsig.2018.06.005.
Kang-Yeoun Jung, Dongsoo Koh, Somi Kim Cho, Yoongho Lim, Quantitative [92] J.-H. Gong, D. Shin, S.-Y. Han, J.-L. Kim, Y.-H. Kang, Kaempferol suppresses
Relationships between the Cytotoxicity of Flavonoids on the Human Breast eosionphil infiltration and airway inflammation in airway epithelial cells and
Cancer Stem-Like Cells MCF7-SC and Their Structural Properties, Chem. Biol. in mice with allergic asthma, J. Nutr. 142 (2012) 47–56, https://doi.org/
Drug Des. 86 (4) (2015) 496–508, https://doi.org/10.1111/cbdd.12512. 10.3945/jn.111.150748.
[70] Fei Ding, Wei Peng, Yu-Kui Peng, Biophysical exploration of protein-flavonol [93] Zuyi Weng, Bodi Zhang, Shahrzad Asadi, Nikolaos Sismanopoulos, Alan
recognition: Effects of molecular properties and conformational flexibility, Butcher, Xueyan Fu, Alexandra Katsarou-Katsari, Christina Antoniou,
Phys. Chem. Chem. Phys. 18 (17) (2016) 11959–11971, https://doi.org/ Theoharis C. Theoharides, Christian Taube, Quercetin is more effective than
10.1039/C5CP07754K. cromolyn in blocking human mast cell cytokine release and inhibits contact
[71] Sam-Yong Park, Takeshi Yokoyama, Naoya Shibayama, Yoshitsugu Shiro, dermatitis and photosensitivity in humans, PLoS ONE 7 (3) (2012) e33805,
Jeremy R.H. Tame, 1.25 Å Resolution Crystal Structures of Human https://doi.org/10.1371/journal.pone.0033805.
Haemoglobin in the Oxy, Deoxy and Carbonmonoxy Forms, J. Mol. Biol. 360 [94] Zuyi Weng, Arti B. Patel, Smaro Panagiotidou, Theoharis C. Theoharides, The
(3) (2006) 690–701, https://doi.org/10.1016/j.jmb.2006.05.036. novel flavone tetramethoxyluteolin is a potent inhibitor of human mast cells,
[72] Richard Bayliss, Teresa Sardon, Isabelle Vernos, Elena Conti, Structural basis J. Allergy Clin. Immunol. 135 (4) (2015) 1044–1052.e5, https://doi.org/
of Aurora-A activation by TPX2 at the mitotic spindle, Mol. Cell. 12 (4) (2003) 10.1016/j.jaci.2014.10.032.
851–862, https://doi.org/10.1016/S1097-2765(03)00392-7. [95] Cristian Del Bo’, Martin Roursgaard, Marisa Porrini, Steffen Loft, Peter Møller,
[73] Kiho Kim, Hyeonjeong Choe, Yujeong Jeong, Jun Hee Lee, Sungwoo Hong, Ru Patrizia Riso, Different effects of anthocyanins and phenolic acids from wild
(II)-Catalyzed Site-Selective Hydroxylation of Flavone and Chromone blueberry (Vaccinium angustifolium) on monocytes adhesion to endothelial
Derivatives: The Importance of the 5-Hydroxyl Motif for the Inhibition of cells in a TNF-a stimulated proinflammatory environment, Mol. Nutr. Food
Aurora Kinases, Org. Lett. 17 (10) (2015) 2550–2553, https://doi.org/10.1021/ Res. 60 (11) (2016) 2355–2366, https://doi.org/10.1002/mnfr.201600178.
acs.orglett.5b01138. [96] Hong-Wei Zhang, Jin-Jiao Hu, Ruo-Qiu Fu, Xin Liu, Yan-Hao Zhang, Jing Li, Lei
[74] R.E. Kartasasmi, R. Herowati, T. Gusdinar, Docking study of quercetin Liu, Yu-Nong Li, Qin Deng, Qing-Song Luo, Qin Ouyang, Ning Gao, Flavonoids
derivatives on inducible Nitric Oxide Synthase and prediction of their inhibit cell proliferation and induce apoptosis and autophagy through
absorption and distribution properties, J. Appl. Sci. 10 (23) (2010) 3098– downregulation of PI3Kc mediated PI3K/AKT/mTOR/p70S6K/ULK signaling
3104, https://doi.org/10.3923/jas.2010.3098.3104. pathway in human breast cancer cells, Sci. Rep. 8 (1) (2018), https://doi.org/
[75] R.J. Rosenfeld, E.D. Garcin, K. Panda, G. Andersson, A. Åberg, A.V. Wallace, G.M. 10.1038/s41598-018-29308-7.
Morris, A.J. Olson, D.J. Stuehr, J.A. Tainer, J.A. Tainer, E.D. Getzoff, Conformational [97] M. Kimata, M. Shichijo, T. Miura, I. Serizawa, N. Inagaki, H. Nagai, Effects of
changes in nitric oxide synthases induced by chlorzoxazone and nitroindazoles: luteolin, quercetin and baicalein on immunoglobulin E- mediated mediator
Crystallographic and computational analyses of inhibitor potency, Biochemistry release from human cultured mast cells, Clin. Exp. Allergy. 30 (2000) 501–
41 (2002) 13915–13925, https://doi.org/10.1021/bi026313j. 508, https://doi.org/10.1046/j.1365-2222.2000.00768.x.
[76] X. Chen, S. Sood, C.S. Yang, N. Li, Z. Sun, Five-lipoxygenase pathway of [98] Alexandre P. Rogerio, Cristiana L. Dora, Edinéia L. Andrade, Juliana S. Chaves,
arachidonic acid metabolism in carcinogenesis and cancer chemoprevention, Luis F.C. Silva, Elenara Lemos-Senna, João B. Calixto, Anti-inflammatory effect

17
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

of quercetin-loaded microemulsion in the airways allergic inflammatory [118] A.K. Mishra, B.K. Singh, A.K. Pandey, In vitro-antibacterial activity and
model in mice, Pharmacol. Res. 61 (4) (2010) 288–297, https://doi.org/ phytochemical profiles of Cinnamomum tamala (Tejpat) leaf extracts and oil,
10.1016/j.phrs.2009.10.005. Rev. Infect. 1 (2010) 134–139.
[99] Wei Duan, I. Chun Kuo, Sathiyamoorthy Selvarajan, Kaw Yan Chua, Boon Huat [119] Rishikesh Prajapati, Santosh Kumar Dubey, Ruchi Gaur, Raj Kumar Koiri,
Bay, W. S. Fred Wong, Antiinflammatory effects of genistein, a tyrosine kinase Brajesh Kumar Maurya, Surendra Kumar Trigun, Lallan Mishra, Structural
inhibitor, on a guinea pig model of asthma, Am. J. Respir. Crit. Care Med. 167 characterization and cytotoxicity studies of ruthenium(II)–dmso–chloro
(2) (2003) 185–192, https://doi.org/10.1164/rccm.200205-420OC. complexes of chalcone and flavone derivatives, Polyhedron 29 (3) (2010)
[100] Caini Liu, Liang Zhu, Koichi Fukuda, Suidong Ouyang, Xing Chen, Chenhui 1055–1061, https://doi.org/10.1016/j.poly.2009.11.012.
Wang, Cun-jin Zhang, Bradley Martin, Chunfang Gu, Luke Qin, Suguna [120] Tarique Hussain, Bie Tan, Ghulam Murtaza, Gang Liu, Najma Rahu,
Rachakonda, Mark Aronica, Jun Qin, Xiaoxia Li, The flavonoid cyanidin blocks Muhammad Saleem Kalhoro, Dildar Hussain Kalhoro, Tolulope O
binding of the cytokine interleukin-17A to the IL-17RA subunit to alleviate Adebowale, Muhammad Usman Mazhar, Zia ur Rehman, Yordan Martínez,
inflammation in vivo, Sci. Signal. 10 (467) (2017) eaaf8823, https://doi.org/ Shahzad Akber Khan, Yulong Yin, Flavonoids and type 2 diabetes: Evidence of
10.1126/scisignal.aaf8823. efficacy in clinical and animal studies and delivery strategies to enhance their
[101] D. Wilson, M. Evans, N. Guthrie, P. Sharma, J. Baisley, F. Schonlau, C. Burki, A therapeutic efficacy, Pharmacol. Res. 152 (2020) 104629, https://doi.org/
randomized, double-blind, placebo-controlled exploratory study to evaluate 10.1016/j.phrs.2020.104629.
the potential of PycnogenolÒ for improving allergic rhinitis symptoms, [121] Ahmad Abdullah, Palaniyandi Ravanan, Kaempferol mitigates Endoplasmic
Phyther. Res. 24 (2010) 1115–1119, https://doi.org/10.1002/ptr.3232. Reticulum Stress Induced Cell Death by targeting caspase 3/7, Sci. Rep. 8 (1)
[102] Amy Rees, Georgina Dodd, Jeremy Spencer, The Effects of Flavonoids on (2018), https://doi.org/10.1038/s41598-018-20499-7.
Cardiovascular Health: A Review of Human Intervention Trials and [122] Yanqing Wu, Fang Wang, E. Albert Reece, Peixin Yang, Curcumin ameliorates
Implications for Cerebrovascular Function, Nutrients. 10 (12) (2018) 1852, high glucose-induced neural tube defects by suppressing cellular stress and
https://doi.org/10.3390/nu10121852. apoptosis, Am. J. Obstet. Gynecol. 212 (6) (2015) 802.e1–802.e8, https://doi.
[103] Arrigo F.G. Cicero, Federica Fogacci, Martina Rosticci, Angelo Parini, Marina org/10.1016/j.ajog.2015.01.017.
Giovannini, Maddalena Veronesi, Sergio D’Addato, Claudio Borghi, Correction [123] M.-J. Goh, J.-S. Park, J.-H. Bae, D.-H. Kim, H.-K. Kim, Y.-J. Na, Effects of ortho-
to: Effect of a short-term dietary supplementation with phytosterols, red dihydroxyisoflavone derivatives from korean fermented soybean paste on
yeast rice or both on lipid pattern in moderately hypercholesterolemic melanogenesis in b16 melanom cells and human skin equivalents, Phyther.
subjects: A three-arm, double-blind, randomized clinical trial (Nutrition and Res. 26 (2012) 1107–1112, https://doi.org/10.1002/ptr.3682.
Metabolism (2017) 14, Nutr. Metab. 15 (1) (2018), https://doi.org/10.1186/ [124] Phil-Dong Moon, Hyung-Mim Kim, Antiinflammatory effects of traditional
s12986-018-0280-0. Korean medicine, JinPi-tang and its active ingredient, hesperidin in HaCaT
[104] Nicola P. Bondonno, Catherine P. Bondonno, Lauren C. Blekkenhorst, Michael cells, Phyther. Res. 26 (5) (2012) 657–662, https://doi.org/10.1002/ptr.3627.
J. Considine, Ghassan Maghzal, Roland Stocker, Richard J. Woodman, Natalie [125] Amani S. Awaad, Reham M. El-Meligy, Gamal A. Soliman, Natural products in
C. Ward, Jonathan M. Hodgson, Kevin D. Croft, Flavonoid-Rich Apple treatment of ulcerative colitis and peptic ulcer, J. Saudi Chem. Soc. 17 (1)
Improves Endothelial Function in Individuals at Risk for Cardiovascular (2013) 101–124, https://doi.org/10.1016/j.jscs.2012.03.002.
Disease: A Randomized Controlled Clinical Trial, Mol. Nutr. Food Res. 62 (3) [126] B.S. Murray, M.V. Babak, C.G. Hartinger, P.J. Dyson, The development of
(2018) 1700674, https://doi.org/10.1002/mnfr.201700674. RAPTA compounds for the treatment of tumors, Coord. Chem. Rev. 306 (2016)
[105] J.G. Gormaz, S. Quintremil, R. Rodrigo, Cardiovascular disease: A target for the 86–114, https://doi.org/10.1016/j.ccr.2015.06.014.
pharmacological effects of quercetin, Curr. Top. Med. Chem. 15 (2015) 1735– [127] Samuel M. Meier-Menches, Christopher Gerner, Walter Berger, Christian G.
1742, https://doi.org/10.2174/1568026615666150427124357. Hartinger, Bernhard K. Keppler, Structure-activity relationships for
[106] Chiemi Kamada, Edson L. da Silva, Mayumi Ohnishi-Kameyama, Jae-Hak ruthenium and osmium anticancer agents-towards clinical development,
Moon, Junji Terao, Attenuation of lipid peroxidation and hyperlipidemia by Chem. Soc. Rev. 47 (3) (2018) 909–928, https://doi.org/10.1039/C7CS00332C.
quercetin glucoside in the aorta of high cholesterol-fed rabbit, Free Radic. [128] Sundarraman Balaji, Mohamed Kasim Mohamed Subarkhan, Rengan Ramesh,
Res. 39 (2) (2005) 185–194, https://doi.org/10.1080/10715760400019638. Hangxiang Wang, David Semeril, Synthesis and Structure of Arene Ru(II)
[107] X. Lin, C.H. Lin, T. Zhao, D. Zuo, Z. Ye, L. Liu, M.T. Lin, Quercetin protects N^O-Chelating Complexes. In Vitro Cytotoxicity and Cancer Cell Death
against heat stroke-induced myocardial injury in male rats: Antioxidative Mechanism, Organometallics 39 (8) (2020) 1366–1375, https://doi.org/
and antiinflammatory mechanisms, Chem. Biol. Interact. 265 (2017) 47–54, 10.1021/acs.organomet.0c00092.
https://doi.org/10.1016/j.cbi.2017.01.006. [129] C. Spoerlein, K. Mahal, H. Schmidt, R. Schobert, Effects of chrysin, apigenin,
[108] Ling Ji, Qiang Du, YunTao Li, Wenzhi Hu, Puerarin inhibits the inflammatory genistein and their homoleptic copper(II) complexes on the growth and
response in atherosclerosis via modulation of the NF-jB pathway in a rabbit metastatic potential of cancer cells, J. Inorg. Biochem. 127 (2013) 107–115,
model, Pharmacol. Reports. 68 (5) (2016) 1054–1059, https://doi.org/ https://doi.org/10.1016/j.jinorgbio.2013.07.038.
10.1016/j.pharep.2016.06.007. [130] K.J. Burke, L.J. Stephens, M.V. Werrett, P.C. Andrews, Bismuth(III)
[109] Y. Zhang, P. Liao, M. Zhu, W. Li, D. Hu, S. Guan, L. Chen, Baicalin Attenuates Flavonolates: The Impact of Structural Diversity on Antibacterial Activity,
Cardiac Dysfunction and Myocardial Remodeling in a Chronic Pressure- Mammalian Cell Viability and Cellular Uptake, Chem. Eur. J. 26 (2020) 7657–
Overload Mice Model, Cell. Physiol. Biochem. 41 (2017) 849–864, https://doi. 7671.
org/10.1159/000459708. [131] Jing-Jing Zhang, Mohamed A. Abu el Maaty, Henrik Hoffmeister, Claudia
[110] Divyashree Ravishankar, Maryam Salamah, Alda Attina, Radhika Pothi, Schmidt, Julienne K. Muenzner, Rainer Schobert, Stefan Wölfl, Ingo Ott, A
Thomas M. Vallance, Muhammad Javed, Harry F. Williams, Eman M.S. Multitarget Gold(I) Complex Induces Cytotoxicity Related to Aneuploidy in
Alzahrani, Elena Kabova, Rajendran Vaiyapuri, Kenneth Shankland, Jonathan HCT-116 Colorectal Carcinoma Cells, Angew. Chemie - Int. Ed. 59 (38) (2020)
Gibbins, Katja Strohfeldt, Francesca Greco, Helen M.I. Osborn, Sakthivel 16795–16800, https://doi.org/10.1002/anie.202006212.
Vaiyapuri, Ruthenium-conjugated chrysin analogues modulate platelet [132] Hana Kostrhunova, Juraj Zajac, Lenka Markova, Viktor Brabec, Jana
activity, thrombus formation and haemostasis with enhanced efficacy, Sci. Kasparkova, A Multi-action Pt IV Conjugate with Oleate and Cinnamate
Rep. 7 (1) (2017), https://doi.org/10.1038/s41598-017-05936-3. Ligands Targets Human Epithelial Growth Factor Receptor HER2 in
[111] T.P. Tim Cushnie, Andrew J. Lamb, Antimicrobial activity of flavonoids, Int. J. Aggressive Breast Cancer Cells, Angew. Chemie. 132 (47) (2020) 21343–
Antimicrob. Agents. 26 (5) (2005) 343–356, https://doi.org/10.1016/j. 21348, https://doi.org/10.1002/ange.202009491.
ijantimicag.2005.09.002. [133] Qian Cao, Dan-Jie Zhou, Zheng-Yin Pan, Gang-Gang Yang, Hang Zhang, Liang-
[112] M.M. Cowan, Plant products as antimicrobial agents, Clin. Microbiol. Rev. 12 Nian Ji, Zong-Wan Mao, CAIXplatins: Highly Potent Platinum(IV) Prodrugs
(4) (1999) 564–582, https://doi.org/10.1128/CMR.12.4.564. Selective Against Carbonic Anhydrase IX for the Treatment of Hypoxic
[113] Ajay K Mishra, Amita Mishra, HK Kehri, Bechan Sharma, Abhay K Pandey, Tumors, Angew. Chemie. 132 (42) (2020) 18715–18721, https://doi.org/
Inhibitory activity of Indian spice plant Cinnamomum zeylanicum extracts 10.1002/ange.202005362.
against Alternaria solani and Curvularia lunata, the pathogenic dematiaceous [134] Patty K.-L. Fu, Patricia M. Bradley, Dietmar van Loyen, Heinz Dürr, Stefan H.
moulds, Ann. Clin. Microbiol. Antimicrob. 8 (1) (2009) 9, https://doi.org/ Bossmann, Claudia Turro, DNA photocleavage by a supramolecular Ru(II)-
10.1186/1476-0711-8-9. viologen complex, Inorg. Chem. 41 (15) (2002) 3808–3810, https://doi.org/
[114] K.A. Ohemeng, C.F. Schwender, K.P. Fu, J.F. Barrett, DNA gyrase inhibitory and 10.1021/ic020136t.
antibacterial activity of some flavones(1), Bioorganic Med. Chem. Lett. 3 (2) [135] M. Tian, J. Li, S. Zhang, L. Guo, X. He, D. Kong, H. Zhang, Z. Liu, Half-sandwich
(1993) 225–230, https://doi.org/10.1016/S0960-894X(01)80881-7. ruthenium(ii) complexes containing N^N-chelated imino-pyridyl ligands
[115] K. Osawa, H. Yasuda, T. Maruyama, H. Morita, K. Takeya, H. Itokawa, that are selectively toxic to cancer cells, Chem. Commun. 53 (2017) 12810–
Isoflavanones from the Heartwood of Swartzia polyphylla and Their 12813, https://doi.org/10.1039/c7cc08270c.
Antibacterial Activity against Cariogenic Bacteria., Chem. Pharm. Bull. [136] R. Li, Y. Ma, X. Hu, W. Wu, X. Wu, C. Dong, S. Shi, Y. Lin, 2+significantly inhibits
(Tokyo). 40 (1992). https://doi.org/10.1248/cpb.40.2970. glioblastoma growth: In vitro and vivo with fewer side-effects than cisplatin,
[116] Hiroyuki Haraguchi, Katsuhito Tanimoto, Yukiyoshi Tamura, Kenji Mizutani, Dalt. Trans. 49 (2020) 8864–8871, https://doi.org/10.1039/d0dt01877e.
Takeshi Kinoshita, Mode of antibacterial action of retrochalcones from [137] Adnan Zahirović, Emira Kahrović, Marina Cindrić, Sandra Kraljević Pavelić,
Glycyrrhiza inflata, Phytochemistry 48 (1) (1998) 125–129, https://doi.org/ Mirsada Hukić, Anja Harej, Emir Turkušić, Heteroleptic ruthenium
10.1016/S0031-9422(97)01105-9. bioflavonoid complexes: from synthesis to in vitro biological activity, J.
[117] Maurya, Surface Functionalization of TiO2 with Plant Extracts and their Coord. Chem. 70 (24) (2017) 4030–4053, https://doi.org/10.1080/
Combined Antimicrobial Activities Against E. faecalis and E, Coli, J. Res. 00958972.2017.1409893.
Updat. Polym. Sci. (2012), https://doi.org/10.6000/1929- [138] L. Mishra, A.K. Singh, Synthesis, spectroscopic, luminescence, electrochemical
5995.2012.01.01.6. and antibacterial studies of ruthenium(II) polypyridyl complexes containing

18
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

3-hydroxyflavones as co-ligand, Indian J. Chem. -Section A. 40A (2001) 1288– carcinoma cells, J. Inorg. Biochem. 105 (4) (2011) 518–524, https://doi.org/
1294. 10.1016/j.jinorgbio.2010.12.013.
[139] Regina M.M. Oliveira, Juliana F. de Souza Daniel, Rose M. Carlos, Synthesis, [156] J. Marques, A.M.S. Silva, M.P.M. Marques, S.S. Braga, Ruthenium(II)
spectroscopic characterization and biological activity of cis-[Ru(hesperidin) trithiacyclononane complexes of 7,30 ,40 -trihydroxyflavone, chrysin and
(1,100 -phenanthroline)2](PF6) complex, J. Mol. Struct. 1031 (2013) 269–274, tectochrysin: Synthesis, characterisation, and cytotoxic evaluation,
https://doi.org/10.1016/j.molstruc.2012.09.066. Inorganica Chim. Acta. 488 (2019) 71–79, https://doi.org/10.1016/j.
[140] Alexandra-Cristina Munteanu, Anna Notaro, Marta Jakubaszek, Joseph ica.2019.01.003.
Cowell, Mickaël Tharaud, Bruno Goud, Valentina Uivarosi, Gilles Gasser, [157] A. Kurzwernhart, W. Kandioller, C. Bartel, S. Bächler, R. Trondl, G.
Synthesis, Characterization, Cytotoxic Activity, and Metabolic Studies of Mühlgassner, M.A. Jakupec, V.B. Arion, D. Marko, B.K. Keppler, C.G.
Ruthenium(II) Polypyridyl Complexes Containing Flavonoid Ligands, Inorg. Hartinger, Targeting the DNA-topoisomerase complex in a double-strike
Chem. 59 (7) (2020) 4424–4434, https://doi.org/10.1021/acs. approach with a topoisomerase inhibiting moiety and covalent DNA binder,
inorgchem.9b03562. Chem. Commun. 48 (2012) 4839–4841, https://doi.org/10.1039/c2cc31040f.
[141] Anna Notaro, Marta Jakubaszek, Nils Rotthowe, Federica Maschietto, Robin [158] Andrea Kurzwernhart, Wolfgang Kandioller, Simone Bächler, Caroline Bartel,
Vinck, Patrick S. Felder, Bruno Goud, Mickaël Tharaud, Ilaria Ciofini, Fethi Sanela Martic, Magdalena Buczkowska, Gerhard Mühlgassner, Michael A.
Bedioui, Rainer F. Winter, Gilles Gasser, Increasing the Cytotoxicity of Ru(II) Jakupec, Heinz-Bernhard Kraatz, Patrick J. Bednarski, Vladimir B. Arion, Doris
Polypyridyl Complexes by Tuning the Electronic Structure of Dioxo Ligands, J. Marko, Bernhard K. Keppler, Christian G. Hartinger, Structure-Activity
Am. Chem. Soc. 142 (13) (2020) 6066–6084, https://doi.org/ relationships of targeted RuII(g 6- P -Cymene) anticancer complexes with
10.1021/jacs.9b12464. flavonol-Derived ligands, J. Med. Chem. 55 (23) (2012) 10512–10522, https://
[142] Anna Notaro, Angelo Frei, Riccardo Rubbiani, Marta Jakubaszek, Uttara Basu, doi.org/10.1021/jm301376a.
Severin Koch, Cristina Mari, Mazzarine Dotou, Olivier Blacque, Jérémie [159] H. Chen, J.A. Parkinson, S. Parsons, R.A. Coxall, R.O. Gould, P.J. Sadler,
Gouyon, Fethi Bedioui, Nils Rotthowe, Rainer F. Winter, Bruno Goud, Stefano Organometallic ruthenium(II) diamine anticancer complexes: Arene-
Ferrari, Mickaël Tharaud, Martina Řezáčová, Jana Humajová, Pavel Tomšík, nucleobase stacking and stereospecific hydrogen-bonding in guanine
Gilles Gasser, Ruthenium(II) Complex Containing a Redox-Active adducts, J. Am. Chem. Soc. 124 (2002) 3064–3082, https://doi.org/
Semiquinonate Ligand as a Potential Chemotherapeutic Agent: From 10.1021/ja017482e.
Synthesis to in Vivo Studies, J. Med. Chem. 63 (10) (2020) 5568–5584, [160] Anna F. A. Peacock, Michael Melchart, Robert J. Deeth, Abraha
https://doi.org/10.1021/acs.jmedchem.0c00431. Habtemariam, Simon Parsons, Peter J. Sadler, Osmium(II) and
[143] Anna Notaro, Marta Jakubaszek, Severin Koch, Riccardo Rubbiani, Orsolya Ruthenium(II) Arene Maltolato Complexes: Rapid Hydrolysis and
Dömötör, Éva A. Enyedy, Mazzarine Dotou, Fethi Bedioui, Mickaël Tharaud, Nucleobase Binding, Chem. - A Eur. J. 13 (9) (2007) 2601–2613,
Bruno Goud, Stefano Ferrari, Enzo Alessio, Gilles Gasser, A Maltol-Containing https://doi.org/10.1002/chem.200601152.
Ruthenium Polypyridyl Complex as a Potential Anticancer Agent, Chem. - A [161] Wolfgang Kandioller, Christian G. Hartinger, Alexey A. Nazarov, Caroline
Eur. J. 26 (22) (2020) 4997–5009, https://doi.org/10.1002/chem.201904877. Bartel, Matthias Skocic, Michael A. Jakupec, Vladimir B. Arion, Bernhard K.
[144] A. Srishailam, N.M. Gabra, Y.P. Kumar, K.L. Reddy, C.S. Devi, D. Anil Kumar, S.S. Keppler, Maltol-derived ruthenium-cymene complexes with tumor
Singh, S. Satyanarayana, Synthesis, characterization; DNA binding and inhibiting properties: The impact of ligand-metal bond stability on
antitumor activity of ruthenium(II) polypyridyl complexes, J. Photochem. anticancer activity in vitro, Chem. - A Eur. J. 15 (45) (2009) 12283–12291,
Photobiol. B Biol. 141 (2014) 47–58, https://doi.org/10.1016/j. https://doi.org/10.1002/chem.200901939.
jphotobiol.2014.09.003. [162] Andrea Kurzwernhart, Stephan Mokesch, Erik Klapproth, Mahsa S. Adib-
[145] L. Gramni, N. Vukea, A. Chakraborty, W.J. Samson, L.M.K. Dingle, B. Xulu, J.A. Ravazi, Michael A. Jakupec, Christian G. Hartinger, Wolfgang Kandioller,
de la Mare, A.L. Edkins, I.N. Booysen, Anticancer evaluation of ruthenium(III) Bernhard K. Keppler, Flavonoid-based organometallics with different metal
complexes with N-donor ligands tethered to coumarin or uracil moieties, centers - Investigations of the effects on reactivity and cytotoxicity, Eur. J.
Inorganica Chim. Acta. 492 (2019) 98–107, https://doi.org/10.1016/j. Inorg. Chem. 2016 (2) (2016) 240–246, https://doi.org/10.1002/
ica.2019.04.018. ejic.201501020.
[146] P. Thangavel, B. Viswanath, S. Kim, Synthesis and characterization of [163] M. Kubanik, J.K.Y. Tu, T. Söhnel, M. Hejl, M.A. Jakupec, W. Kandioller, B.K.
kaempferol-based ruthenium (II) complex: A facile approach for superior Keppler, C.G. Hartinger, Expanding on the Structural Diversity of Flavone-
anticancer application, Mater. Sci. Eng. C. 89 (2018) 87–94, https://doi.org/ Derived RutheniumII(ƞ6-arene) Anticancer Agents, Metallodrugs. 1 (2016),
10.1016/j.msec.2018.03.020. https://doi.org/10.1515/medr-2015-0001.
[147] B.A. Lakshmi, J.Y. Bae, J.H. An, S. Kim, Facile design and spectroscopic [164] Andrea Kurzwernhart, Wolfgang Kandioller, Éva A. Enyedy, Maria Novak,
characterization of novel bio-inspired Quercetin-conjugated tetrakis Michael A. Jakupec, Bernhard K. Keppler, Christian G. Hartinger, 3-
(dimethylsulfoxide)dichlororuthenium(II) complex for enhanced anticancer Hydroxyflavones vs. 3-hydroxyquinolinones: Structure-activity
properties, Inorganica Chim. Acta. 495 (2019) 118989-undefined. https://doi. relationships and stability studies on RuII(arene) anticancer complexes
org/10.1016/j.ica.2019.118989. with biologically active ligands, Dalt. Trans. 42 (17) (2013) 6193–6202,
[148] R. Gaur, L. Mishra, Bi-nuclear Ru(ii) complexes of bis-chalcone and bis- https://doi.org/10.1039/C2DT32206D.
flavonol: synthesis, characterization, photo cleavage of DNA and [165] S. Movassaghi, E. Leung, M. Hanif, B.Y.T. Lee, H.U. Holtkamp, J.K.Y. Tu, T.
Topoisomerase I inhibition, RSC Adv. 3 (2013) 12210–12219, https://doi. Söhnel, S.M.F. Jamieson, C.G. Hartinger, A Bioactive l -Phenylalanine-Derived
org/10.1039/c3ra41451e. Arene in Multitargeted Organoruthenium Compounds: Impact on the
[149] L. Mishra, A.K. Singh, Synthesis, spectroscopic, luminescence, electrochemical Antiproliferative Activity and Mode of Action, Inorg. Chem. 57 (2018)
and antitumour/anti-HIV studies of some new monomeric and dimeric 8521–8529, https://doi.org/10.1021/acs.inorgchem.8b01187.
ruthenium(II) complexes of 3-hydroxy-4’-benzyloxy flavone containing [166] Carmen M. Hackl, Beatrix Schoenhacker-Alte, Matthias H.M. Klose, Helena
nitrogen, oxygen and sulphur donors as co-ligands, Indian J. Chem. 41A Henke, Maria S. Legina, Michael A. Jakupec, Walter Berger, Bernhard K.
(2002) 1826–1831. Keppler, Oliver Brüggemann, Ian Teasdale, Petra Heffeter, Wolfgang
[150] Souvik Roy, Anweshan Sil, Tania Chakraborty, Potentiating apoptosis and Kandioller, Synthesis and: In vivo anticancer evaluation of poly(organo)
modulation of p53, Bcl2, and Bax by a novel chrysin ruthenium complex for phosphazene-based metallodrug conjugates, Dalt. Trans. 46 (36) (2017)
effective chemotherapeutic efficacy against breast cancer, J. Cell. Physiol. 234 12114–12124, https://doi.org/10.1039/C7DT01767G.
(4) (2019) 4888–4909, https://doi.org/10.1002/jcp.27287. [167] Sushma L. Saraf, Trevor J. Fish, Abby D. Benninghoff, Ashley A. Buelt, Rhett C.
[151] Yixuan Wang, Li Bian, Tania Chakraborty, Torsha Ghosh, Pallakhi Chanda, Smith, Lisa M. Berreau, Photochemical reactivity of RuII(g6-p-cymene)
Souvik Roy, Construing the Biochemical and Molecular Mechanism flavonolato compounds, Organometallics 33 (22) (2014) 6341–6351,
Underlying the In Vivo and In Vitro Chemotherapeutic Efficacy of https://doi.org/10.1021/om5006337.
Ruthenium-Baicalein Complex in Colon Cancer, Int. J. Biol. Sci. 15 (5) [168] Xiaozhen Han, Kevin K. Klausmeyer, Patrick J. Farmer, Characterization of the
(2019) 1052–1071, https://doi.org/10.7150/ijbs.31143. Initial Intermediate Formed during Photoinduced Oxygenation of the
[152] Ashok Kumar Singh, Gunjan Saxena, Sahabjada, M. Arshad, Synthesis, Ruthenium(II) Bis(bipyridyl)flavonolate Complex, Inorg. Chem. 55 (15)
characterization and biological evaluation of ruthenium flavanol complexes (2016) 7320–7322, https://doi.org/10.1021/acs.inorgchem.6b00852.
against breast cancer, Spectrochim. Acta - Part A Mol. Biomol. Spectrosc. 180 [169] Xiaozhen Han, Murugaeson R. Kumar, Amanda Hoogerbrugge, Kevin K.
(2017) 97–104, https://doi.org/10.1016/j.saa.2017.02.056. Klausmeyer, Mukunda M. Ghimire, Lauren M. Harris, Mohammad A. Omary,
[153] J. Ochocki, M. Kasprzak, L. Chȩcińska, A. Erxleben, E. Zyner, L. Szmigiero, A. Patrick J. Farmer, Mechanistic Investigations of Photoinduced Oxygenation of
Garza-Ortiz, J. Reedijk, Synthesis, single-crystal and solution structure Ru(II) Bis-bipyridyl Flavonolate Complexes, Inorg. Chem. 57 (5) (2018) 2416–
analysis and in vitro cytotoxic activity of two novel complexes of 2424, https://doi.org/10.1021/acs.inorgchem.7b01384.
ruthenium(ii) with in situ formed flavanone-based ligands, Dalt. Trans. 39 [170] Wolfgang Kandioller, Christian G. Hartinger, Alexey A. Nazarov, Maxim L.
(2010) 9711–9718, https://doi.org/10.1039/c0dt00535e. Kuznetsov, Roland O. John, Caroline Bartel, Michael A. Jakupec, Vladimir B.
[154] M. Kasprzak, M. Fabijańska, L. Che˛cińska, K. Studzian, M. Markowicz- Arion, Bernhard K. Keppler, From pyrone to thiopyrone ligands-rendering
Piasecka, J. Sikora, E. Mikiciuk-Olasik, J. Ochocki, Small differences in maltol-derived ruthenium(II)-arene complexes that are anticancer active
structure, a large difference in activity – Comparing a new Ru(II)-3- in vitro, Organometallics 28 (15) (2009) 4249–4251, https://doi.org/10.1021/
hydroxyiminoflavanone complex with analogous Ru(II) compounds, om900483t.
Inorganica Chim. Acta. 457 (2017) 69–80, https://doi.org/10.1016/j. [171] Wolfgang Kandioller, Andrea Kurzwernhart, Muhammad Hanif, Samuel M.
ica.2016.11.021. Meier, Helena Henke, Bernhard K. Keppler, Christian G. Hartinger, Pyrone
[155] Maria M. Kasprzak, Leszek Szmigiero, Elzbieta _ Zyner, Justyn Ochocki, derivatives and metals: From natural products to metal-based drugs, J.
Proapoptotic activity in vitro of two novel ruthenium(II) complexes with Organomet. Chem. 696 (5) (2011) 999–1010, https://doi.org/10.1016/j.
flavanone-based ligands that overcome cisplatin resistance in human bladder jorganchem.2010.11.010.

19
M. Małecka, A. Skoczyńska, D.M. Goodman et al. Coordination Chemistry Reviews 436 (2021) 213849

[172] Shipra Yadav, Jai Deo Singh, Synthesis and preliminary biological evaluation Czyz, Tautomerization of 2-nitroso-N-arylanilines by coordination as N, N’-
for the anticancer activity of organochalcogen (S/se) tethered chrysin-based chelate ligands to rhenium(i) complexes and the anticancer activity of newly
organometallic RuII(g6-p-cymene) complexes, J. Biomol. Struct. Dyn. 37 (13) synthesized oximine rhenium(i) complexes against human melanoma and
(2019) 3337–3353, https://doi.org/10.1080/07391102.2018.1513867. leukemia cells in vitro, J. Inorg. Biochem. 104 (7) (2010) 774–789, https://doi.
[173] Erin E. Battin, Julia L. Brumaghim, Antioxidant activity of sulfur and org/10.1016/j.jinorgbio.2010.03.014.
selenium: A review of reactive oxygen species scavenging, glutathione [176] A. Pastuszko, K. Majchrzak, M. Czyz, B. Kupcewicz, E. Budzisz, The synthesis,
peroxidase, and metal-binding antioxidant mechanisms, Cell Biochem. lipophilicity and cytotoxic effects of new ruthenium(II) arene complexes with
Biophys. 55 (1) (2009) 1–23, https://doi.org/10.1007/s12013-009-9054-7. chromone derivatives, J. Inorg. Biochem. 159 (2016) 133–141, https://doi.org/
[174] Adam Pastuszko, Karolina Niewinna, Malgorzata Czyz, Andrzej Jóźwiak, 10.1016/j.jinorgbio.2016.02.020.
Magdalena Małecka, Elzbieta Budzisz, Synthesis, X-ray structure, [177] P. Mucha, P. Hikisz, K. Gwoździński, U. Krajewska, A. Leniart, E. Budzisz,
electrochemical properties and cytotoxic effects of new arene ruthenium(II) Cytotoxic effect, generation of reactive oxygen/nitrogen species and
complexes, J. Organomet. Chem. 745-746 (2013) 64–70, https://doi.org/ electrochemical properties of Cu(ii) complexes in comparison to half-
10.1016/j.jorganchem.2013.07.020. sandwich complexes of Ru(ii) with aminochromone derivatives, RSC Adv. 9
[175] Stefan Wirth, Andreas U. Wallek, Anna Zernickel, Florian Feil, M. Sztiller- (2019) 31943–31952, https://doi.org/10.1039/c9ra05971g.
Sikorska, K. Lesiak-Mieczkowska, Christoph Bräuchle, Ingo-Peter Lorenz, M.

20

You might also like