Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

pharmaceuticals

Review
Review
Monofunctional Platinum(II)
Monofunctional Platinum(II) Anticancer
Anticancer Agents
Agents
Suxing Jin
Suxing Jin1,†
1,†, Yan Guo 1,2,†1,2,†
, Yan Guo , Zijian GuoGuo
, Zijian
1 and Xiaoyong
1 and Wang
Xiaoyong
3,* 3,
Wang *

1 State
StateKey
KeyLaboratory
LaboratoryofofCoordination
CoordinationChemistry,
Chemistry, School
School of
of Chemistry
Chemistry and and Chemical
Chemical Engineering,
Engineering, Nanjing
University, Nanjing 210023,
Nanjing University, NanjingChina; jinsuxing@nju.edu.cn
210023, (S.J.); 20191022@hncj.edu.cn
China; jinsuxing@nju.edu.cn (Y.G.); (Y.G.);
(S.J.); 20191022@hncj.edu.cn
zguo@nju.edu.cn
zguo@nju.edu.cn(Z.G.)
(Z.G.)
22 School
Schoolof
ofMaterials
Materialsand
andChemical
ChemicalEngineering,
Engineering,Henan
Henan University
University of of Urban
Urban Construction,
Construction, Pingdingshan
467036, China 467036, China
Pingdingshan
33 State
StateKey
KeyLaboratory
LaboratoryofofPharmaceutical
PharmaceuticalBiotechnology,
Biotechnology, School
School of of Life
Life Sciences,
Sciences, Nanjing
Nanjing University,
University, Nanjing
210023, China
Nanjing 210023, China
** Correspondence:
Correspondence:boxwxy@nju.edu.cn
boxwxy@nju.edu.cn
†† These
Theseauthors
authorscontributed
contributedequally
equallyto
tothis
thisarticle.
article.

Abstract: Platinum-based
Abstract: Platinum-based anticancer
anticancer drugs
drugs represented
represented by by cisplatin
cisplatin play
play important
important roles
roles inin the
the
treatment of
treatment of various
various solid
solid tumors.
tumors. However,
However, their
their applications
applications are are largely
largely compromised
compromised by by drug
drug
resistanceand
resistance andside
side effects.
effects. Much
Much effort
effort has
has been
been made
made to to circumvent
circumvent the the drug
drug resistance
resistanceand
and general
general
toxicity of these drugs. Among multifarious designs, monofunctional platinum(II)
toxicity of these drugs. Among multifarious designs, monofunctional platinum(II) complexes with complexes with
aa general
general formula
formula of of [Pt(3A)Cl]
[Pt(3A)Cl] (A: (A: Ammonia
Ammonia or or amine)
amine) stand
stand out as aa class
class ofof “non-traditional”
“non-traditional”
+
+ out as
anticancer agents
anticancer agents hopeful
hopeful to to overcome
overcome the the defects
defects ofof current
current platinum
platinum drugs.
drugs. This
This review
review aims
aims toto
summarize the development of monofunctional platinum(II) complexes
summarize the development of monofunctional platinum(II) complexes in recent years. They are in recent years. They are
classified into
classified into four
four categories: fluorescent complexes,
categories: fluorescent complexes, photoactive
photoactive complexes,
complexes, targeted
targeted complexes,
complexes,
and miscellaneous
and miscellaneous complexes.
complexes. The intention behind
The intention behind thethe designs
designs isis either
either toto visualize
visualize the
the cellular
cellular
distribution, or
distribution, or to
to reduce
reduce thethe side
side effects,
effects, or
or to
to improve
improve the the tumor
tumor selectivity,
selectivity, or or inhibit
inhibit the
the cancer
cancer
cells through non-DNA targets. The information provided by this review
cells through non-DNA targets. The information provided by this review may inspire researchers tomay inspire researchers
to conceive
conceive moremore innovative
innovative complexes
complexes with
with potentefficacy
potent efficacytotoshake
shakeoffoffthe
thedrawbacks
drawbacksof ofplatinum
platinum

 anticancerdrugs.
anticancer drugs.
Citation: Jin, S.; Guo, Y.; Guo, Z.;
Citation: Jin, S.; Guo, Y.; Guo, Z.;
Wang, X. Monofunctional Keywords: anticancer
anticancer drug;
drug; drug
drug design;
design; metal-based
metal-baseddrug;
drug;monofunctional
monofunctionalplatinum
platinumcomplex
complex
Wang, X. Monofunctional Keywords:
Platinum(II) Anticancer Agents.
Platinum(II) Anticancer Agents.
Pharmaceuticals 2021, 14, x.
Pharmaceuticals 2021, 14, 133.
https://doi.org/10.3390/xxxxx
https://doi.org/10.3390/ph14020133
1. Introduction
1. Introduction
Academic Editor: Guido Crisponi
Academic Editor: Guido Crisponi
Received: 12 January 2021 Cisplatin and
Cisplatin and its
its analogues,
analogues, carboplatin,
carboplatin, oxaliplatin,
oxaliplatin, nedaplatin [1], lobaplatin [2],
Received: 12 January 2021
Accepted: 4 February 2021 and heptaplatin
and heptaplatin [3] (Figure 1) 1) have
havebeen
beenapproved
approvedfor forclinical
clinicaluse
useinindifferent
different countries
countries to
Accepted: 4 February 2021
Published: 7 February 2021 to treat
treat multiplesolid
multiple solidneoplasms,
neoplasms,and andapproximately
approximatelyhalf half of
of the chemotherapy
chemotherapy strategies
strategies
Published: 7 February 2021
include
include platinum
platinum drugs
drugs [4–6].
[4–6]. However,
However, these
these drugs
drugs are
are structural
structural congeners
congeners of of cisplatin
cisplatin
Publisher’s Note: MDPI stays neu- and
and therefore, some drawbacks are inherited [7–9]. For instance, DNA is believed to
therefore, some drawbacks are inherited [7–9]. For instance, DNA is believed to be
be
Publisher’s Note: MDPI stays neutral
tral with regard to jurisdictional the ultimate target of cisplatin, so are other platinum drugs [10–12]. Nevertheless,
the ultimate target of cisplatin, so are other platinum drugs [10–12]. Nevertheless, DNA DNA
with regard to jurisdictional claims in
claims in published maps and institu- damages
published maps and institutional affil- damages could
could bebe easily
easily repaired
repaired by
by DNA
DNA repair
repairmechanisms.
mechanisms. Therefore,
Therefore, all
all the
the existing
existing
tional affiliations. platinum
iations. platinum anticancer
anticancer drugs
drugs encounter
encounter drug
drug resistance
resistance [13,14].
[13,14]. Moreover,
Moreover, their
their nonspecific
nonspecific
accumulation
accumulation in in the
thehypermetabolic
hypermetabolicstate
statetissues
tissuesresults
resultsininthe
thesystemic
systemictoxicity.
toxicity.

Copyright: © 2021 by the authors. Li-


censee MDPI,
Copyright: Basel,
© 2021 by Switzerland.
the authors.
This article
Licensee is an Basel,
MDPI, open access article
Switzerland.
distributed
This article under the terms
is an open and
access con-
article
ditions of theunder
distributed Creative
theCommons At-
terms and
tribution (CC
conditions BY) Creative
of the license (http://crea-
Commons
tivecommons.org/licenses/by/4.0/).
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/). Figure 1.
Figure 1. Chemical
Chemical structures
structures of
of clinically
clinically approved
approved platinum
platinum anticancer
anticancer drugs.
drugs.

Pharmaceuticals 2021, 14, x. https://doi.org/10.3390/xxxxx www.mdpi.com/journal/pharmaceuticals


Pharmaceuticals 2021, 14, 133. https://doi.org/10.3390/ph14020133 https://www.mdpi.com/journal/pharmaceuticals
ture-activity relationships (SAR) of platinum complexes [15]. Their an
arises from diverse interactions with different biomolecules and disp
trum of activity in favor of circumventing the drug resistance or side e
Pharmaceuticals 2021, 14, 133 2 of 23
DNA remains the major, if not the only, target for most of mono
plexes. Nevertheless, the DNA-binding mode or process is different f
and Oneitsstrategy
analogues. Each
for increasing themonofunctional
potency while mitigating complex could
the side effects form at mos
of platinum
complexes
with the is to exploit new compounds
N7-guanine on the DNA that operate on novel
strands mechanisms
rather than[15,16]. In this
two covalent Pt
respect, cationic monofunctional PtII complexes that contain only one labile ligand exhibit
cisplatin
special doesactivities
anticancer [23]. The earliestwith
in comparison prototype complexes
cisplatin analogues due to[Pt(NH
the changes3)3Cl]+ an
in=DNA-binding
diethylenetriamine) are thought
mode, cellular accumulation, and toeven
bethe inactive
mechanism towards cancer cells
of action [17,18].
These complexes represent an alternative class of anticancer agents that violateIIthe classical
the prevailing view only neutral and square-planar Pt complexes wi
structure-activity relationships (SAR) of platinum complexes [15]. Their antineoplastic
ands arises
activity in afrom cis-configuration
diverse interactions with possess
different anticancer
biomolecules and activity
displays a[6,24,25].
distinct Ho
spectrum of activity in favor of circumventing the drug resistance or side effects [19–22].
ceived belief was overturned by the finding that cis-[Pt(NHII3)2(Am)Cl]
DNA remains the major, if not the only, target for most of monofunctional Pt com-
N-heterocyclic
plexes. Nevertheless, the amine) inhibited
DNA-binding mode ortumorprocess is cells in from
different vitrothatand leukemia (
of cisplatin
and
mouseits analogues.
models Each monofunctional
[26], where Ptcomplex II formed couldstable
form at mostPt–DNA one covalent bond and t
adducts
with the N7-guanine on the DNA strands rather than two covalent Pt−DNA cross-links as
lated into
cisplatin DNA.
does [23]. Further
The earliest studies
prototype found
complexes that
[Pt(NH amino
+ or Am groups
3 )3 Cl] and [Pt(dien)Cl] (dien
+ coul
=to DNA, thereby
diethylenetriamine) are achieving a bifunctional
thought to be inactive towards cancer coordination [27].to the
cells, since according
prevailing view only neutral and square-planar PtII complexes with a pairIIof inert ligands
Afterwards, it was found that the cationic Pt complex pyripl
in a cis-configuration possess anticancer activity [6,24,25]. However, the preconceived
formed
belief a monofunctional
was overturned by the findingadduct with3 )DNA
that cis-[Pt(NH 2 (Am)Cl] and
+ (Am induced littleN-distort
is an aromatic
heterocyclic amine) inhibited tumor cells in vitro and leukemia (L1210 and P388) in mouse
ble helix upon binding. In addition, organic cation transporters (OCTs)
models [26], where PtII formed stable Pt–DNA adducts and the complex intercalated into
cellular
DNA. Further uptake and activity
studies found that amino [28,29].
or Am groups SAR couldstudies
lose uponindicated
binding to thatDNA,the ste
thereby achieving a bifunctional coordination [27].
pyridine ligand played an important IIrole in regulating the action of RN
Afterwards, it was found that the cationic Pt complex pyriplatin (Figure 2) only
formedPhenanthriplatin
32]. a monofunctional adduct (Figure
with DNA 2) alsoand formed
induced little monofunctional
distortion in the DNA adducts w
well helix
double as duplex DNAInonce
upon binding. theorganic
addition, chloridecationligand
transporters lost. Phenanthriplatin-DN
(OCTs) were involved
in its cellular uptake and activity [28,29]. SAR studies indicated that the steric hindrance of
steric hindrance in the major groove of DNA and thus, stall the progr
the pyridine ligand played an important role in regulating the action of RNA polymerase
IImerase II on the damaged
[30–32]. Phenanthriplatin (Figure 2)DNA templates
also formed and inhibit
monofunctional adductsDNA polymeras
with guanine
bases as well as duplex DNA once the chloride ligand lost. Phenanthriplatin-DNA adducts
has a unique cytotoxic profile as it was 7–40 times more active than cisp
generate steric hindrance in the major groove of DNA and thus, stall the progression of
cancer
RNA cell lines
polymerase and,
II on the unlike
damaged DNApyriplatin,
templates and showed
inhibit DNA an activity
polymerases spectrum
[33]. in
This complexpanel
NCI-60 has a unique cytotoxic
of cell linesprofile as it was
[34,35]. 7–40 times
Other more active than cisplatin
monofunctional PtII comple
in many human cancer cell lines and, unlike pyriplatin, showed an activity spectrum in
RNA
the more polymerase
extensive NCI-60IIpanel andofDNA cell linessynthesis,
[34,35]. Other and displayedPthigh
monofunctional cytotoxicit
II complexes

were
that also reported
suppressed [36–39].
RNA polymerase Yet,
II and DNA this is notand
synthesis, thedisplayed
whole story on the mecha
high cytotoxicity
against cancer cells were also reported [36–39]. Yet, this is not the whole story on the
and more studies revealed that the target of monofunctional PtII compl
mechanism of action. More and more studies revealed that the target of monofunctional
Ptor even relevant
II complexes to DNA.
is not limited to or even Therefore,
relevant to DNA. the Therefore,
established SARs no
the established SARs longer f
no longer fit them. This review will introduce some representative monofunctional PtII
will introduce some representative monofunctional Pt complexes pu II
complexes published in the past 5 years or so and discuss their mechanism of action
ifyears
possible. or so and discuss their mechanism of action if possible.

Figure
Figure2. Chemical structuresstructures
2. Chemical of pyriplatin and phenanthriplatin.
of pyriplatin and phenanthriplatin.

2. Fluorescent Monofunctional PtII Complexes


DNA has been extensively studied as the ultimate cellular targ
plexes, while much of the mechanism of action still remains unkno
Pharmaceuticals 2021, 14, 133 3 of 23

2. Fluorescent Monofunctional PtII Complexes


Pharmaceuticals 2021, 14, x FOR PEER REVIEW
DNA has been extensively studied as the ultimate cellular target of platinum 3com- of 24

plexes, while much of the mechanism of action still remains unknown. Although some
small ions or molecules, amino acids, peptides, and proteins are thought to be implicated
in
in the
the mechanism,
mechanism,the thedetails
detailsononthe
thecellular
cellularinteractions
interactionsare largely
are largely unclear.
unclear. Therefore,
Therefore, it isit
of great significance to study the behavior of Pt II complexes in cancer cells at the molecular
is of great significance to study the behavior of Pt complexes in cancer cells at the molec-
II

level. Tethering fluorophores to the II center


ular level. Tethering fluorophores toPtthe of the
PtII center of complexes
the complexes could formform
could fluorescent
fluores-
molecules, by thisbymeans
cent molecules, the cellular
this means locationlocation
the cellular of the complexes could becould
of the complexes mapped be through
mapped
fluorescence imaging. imaging.
through fluorescence
Dinuclear II
Dinuclear PtPtII complex
complex 11 incorporate
incorporate aa fluorescent
fluorescent anthraquinone
anthraquinone intercalator
intercalator in in the
the
structure (Figure 3). Its major merit is to monitor the subcellular localization
structure (Figure 3). Its major merit is to monitor the subcellular localization by fluores- by fluorescence
microscopy on account
cence microscopy of the innate
on account fluorescence
of the innate of the intercalator.
fluorescence Complex
of the intercalator. 1 exhibited
Complex 1 ex-
high
hibited high cytotoxicity in the U2-OS cell line (the designation of U2-OS and cell
cytotoxicity in the U2-OS cell line (the designation of U2-OS and those of other lines
those of
appeared
other cellhereafter are listed
lines appeared in Appendix
hereafter A Table
are listed in A1 at the end
Appendix A of the article)
Table A1 at theandendovercame
of the
resistance
article) andinovercame
the cisplatin-resistant
resistance in the U2-OS/Pt cell line. Their
cisplatin-resistant cellular
U2-OS/Pt process
cell in both
line. Their cell
cellular
lines was similar, which may be due to the formation of intercalative
process in both cell lines was similar, which may be due to the formation of intercalative DNA-adducts that
could evade thethat
DNA-adducts DNA repair
could mechanism
evade the DNA responsible for removing
repair mechanism the cisplatin
responsible adducts [40].
for removing the
The fluorescence indicated that 1 rapidly entered the U2-OS
cisplatin adducts [40]. The fluorescence indicated that 1 rapidly entered the U2-OS cellscells and accumulated in
the nucleus, thereby reaching the biological target of the Pt and
and accumulated in the nucleus, thereby reaching the biological target of the Pt and inter-intercalating moieties—
DNA [41,42].
calating The Pt moiety
moieties—DNA wasThe
[41,42]. excreted fromwas
Pt moiety the cell via the
excreted Golgi
from the apparatus,
cell via the while
Golgi
the weakly basic anthraquinone ligand accumulated in the Golgi complex, where it was
apparatus, while the weakly basic anthraquinone ligand accumulated in the Golgi com-
taken up by lysosomes and then transported to the cell surface. Interestingly, contrasting
plex, where it was taken up by lysosomes and then transported to the cell surface. Inter-
results were found in A2780 cells, implying that different cell lines may respond to Pt drugs
estingly, contrasting results were found in A2780 cells, implying that different cell lines
differently [41]. In cisplatin-resistant A2780 cells, the complexes were sequestered into
may respond to Pt drugs differently [41]. In cisplatin-resistant A2780 cells, the complexes
lysosomes and displayed cross-resistance with cisplatin.
were sequestered into lysosomes and displayed cross-resistance with cisplatin.

Figure 3.
Figure 3. Chemical structures
structures of
of complexes
complexes 1–5.
1‒5.

Fluorescent complexes 22and


Fluorescent complexes and33(Figure
(Figure3)3)were
wereused
usedtoto track
track their
their cellular
cellular distribu-
distribution
tion via detecting the fluorescence, thus providing new insights into the
via detecting the fluorescence, thus providing new insights into the mechanism of actionmechanism of
action [43]. Complex 3 is more suitable for cellular imaging than 2. Particularly, in
[43]. Complex 3 is more suitable for cellular imaging than 2. Particularly, in contrast to the contrast
to the rapid
rapid entry entry to but
to cells cellsinaccessibility
but inaccessibility
to thetonucleus
the nucleus for ligand,
for the the ligand, cationic
cationic 3 entered
3 entered the
the
HeLaHeLa cells
cells slowly
slowly and
and mainly
mainly accumulatedininthe
accumulated thenucleoli.
nucleoli.ItItbound
bound toto the
the cytoplasmic
cytoplasmic
vacuoles, resulting in a different distribution pattern from that of neutral fluorescent Pt
complexes. Complex 3 not only acted as a probe to trace its cellular behavior, but also
induced non-apoptotic cell death. Similarly, complex 4 (Figure 3) realized the in vitro and
vivo fluorescence imaging [44]. Its cellular uptake was much slower than that of the ligand
Pharmaceuticals 2021, 14, 133 4 of 23

vacuoles, resulting in a different distribution pattern from that of neutral fluorescent Pt


complexes. Complex 3 not only acted as a probe to trace its cellular behavior, but also
induced non-apoptotic cell death. Similarly, complex 4 (Figure 3) realized the in vitro and
vivo fluorescence imaging [44]. Its cellular uptake was much slower than that of the ligand
and it could get into the nucleus, suggesting that the PtII center played an important role in
reducing the uptake process and promoting its affinity for DNA. Complex 4 also exhibited
preferential affinity for mitochondria.
Another cytotoxic fluorescent complex 5 (Figure 3) constructed by tethering a fluo-
rophore thioflavin-T (ThT) derivative to the 3N-chelated PtII center was suitable for cellular
imaging in living cells [45]. Fluorescence imaging showed that 5 was sequestrated in
mitochondria and acidic lysosomes after slow entry to the cells. The finding provides new
insights into the cellular distribution of positively-charged monofunctional PtII complexes.
It should be noted that in analyzing the distribution of fluorescent complexes, a comparison
with the distribution of free ligands is necessary so as to eliminate the false fluorescence
emitted by the dissociated fluorophores.

3. Photoactive Monofunctional PtII Complexes


The toxic side effects resulted from the non-specific accumulation of Pt anticancer
agents hinder their broader application in clinical treatment. Photoactive Pt complexes offer
an opportunity to develop new anticancer drugs responsive to light [46,47]. Photodynamic
therapy (PDT) is a minimally invasive method that produces selective cytotoxicity to
malignant tumor cells. It has been used to treat different tumors, such as bladder and
prostate cancers [48]. The anticancer effect of PDT is achieved by killing cancer cells through
reactive oxygen species (ROS) such as singlet oxygen (1 O2 ) produced by photosensitizers
(PSs) under light irradiation [49]. Non-irradiated PSs generally have low dark toxicity, high
1 O quantum yield, and good cellular accessibility [50], while the irradiated one is strongly
2
bioactive. It has been shown that the combination of Pt complexes with PSs could produce
synergistic effects [6,51,52].
Porphyrins are representative PSs for PDT owing to the ring structure with 18 con-
jugated π electrons, which endow the compounds with photophysical properties and
selective retention or accumulation in tumors due to preferential binding to low density
lipoproteins [53,54]. The structure of porphyrin is susceptible to functionalization of sur-
rounding substituents, especially the presence of metal complexes around or in the core
could modify the local environment, improving the solubility or introducing specific func-
tionalities [54,55]. In order to potentiate the action of these structures, conjugations with
PtII complexes were tried. The isomeric tetra-cationic(pyridyl)porphyrin PtII complexes 6
and 7 (Figure 4) possessed cytotoxicity against metastatic WM1366 cells under white-light
irradiation, inducing apoptosis via the activation of caspase-3 and -9 and alteration of cell
cytoskeleton actin [56]. In silico study indicated that these complexes could be employed
to deliver drugs owing to the affinity to the N-terminal region of ApoB-100.
Complex 8 (Figure 4) showed an excellent photocytotoxicity (50 W LED light, 6 J cm−2 ,
30 min) due to the high 1 O2 quantum yield, nuclear internalization, and a caspase-3-
induced apoptosis with negligible dark toxicity [57]. Oxaliplatin was chosen as the phar-
macophore since its DNA binding rate is faster than that of cisplatin. Moreover, it could
improve the hydrophilicity and eliminate the formation of aggregates by increasing the
electrostatic repulsion from charged functional groups [58]. Furthermore, 8 completely
wiped out the tumor tissue in colon26 tumor-bearing mice. Similarly, the aqueous solubil-
ity, cellular uptake, and photophysical property of the tetraplatinated porphyrin complex
9 (Figure 4) were improved by incorporating PtII moieties [51], which directed the por-
phyrin to the nucleus and enhanced the nuclear Pt accumulation. The binding to DNA
involves both covalent bonding with N7-guanine by PtII and intercalation by the porphyrin
unit. Complex 9 demonstrated a promising photocytotoxicity with extremely high toxicity
towards human cancer cell lines upon irradiation (6.95 J cm−2 , 420 nm, 15 min, HeLa:
Pharmaceuticals 2021, 14, 133 5 of 23

ceuticals 2021, 14, x FOR PEER REVIEW IC50 = 37 nM; A2780: IC50 = 21 nM; CP70: IC50 = 19 nM), and a phototoxic
5 of 24 index up to
5000 in the cisplatin-resistant CP70 cell line.

Figure 4. Chemical structures


Figureof4.complexes
Chemical6‒9.
structures of complexes 6–9.

Despite the fact Despite the fact that porphyrin-based


that porphyrin-based Pt complexes have Pt complexes have manythe
many advantages, advantages,
poor the poor
solubility and solubility
aggregation andofaggregation
porphyrinsofaffect porphyrins affect their
their cellular uptake cellular
and uptake andactiv-
limit their limit their activities
and applications
ities and applications in vivo. Thus,in vivo. Thus, water-soluble
water-soluble tumor-targeted tumor-targeted
PS 10 (FigurePS 5) 10
with (Figure
a 5) with a
porphyrin
porphyrin framework framework
containing Gacontaining GaIII andwas
III and PtII moieties PtIIdeveloped
moieties was developed
[59]. Complex[59]. 10 Complex 10
is an efficient 1isOan efficient 1owing
2 generator O2 generator owing atom
to the heavy to theeffect,
heavy acidic
atom effect,
pKa, andacidic pKa, and localization in
localization
in cytosol. It showed negative dark cytotoxicity due to the larger hydrophilicity, slower slower and
cytosol. It showed negative dark cytotoxicity due to the larger hydrophilicity,
lower
and lower cellular cellular
uptake. uptake. Moreover,
Moreover, it exhibited it exhibited
remarkable remarkable photocytotoxicity
photocytotoxicity and inter-and interaction
action with DNA,withaccumulated
DNA, accumulatedin tumorinprominently
tumor prominently (tumor/muscle
(tumor/muscle ratioinhib-
ratio > 9), and > 9), and inhibited
ited tumor growthtumoralmost
growth almost completely
completely over 2No
over 2 weeks. weeks. No significant
significant systemicsystemic
toxicitytoxicity
in- including
cluding weightweightloss andlossadverse
and adverse
reactions reactions were observed.
were observed.
The low efficiencyThe lowof 1Oefficiency
2 generation of 1within
O2 generation
the maximumwithintissue
the maximum
penetrating tissue
and bi- penetrating and
ocompatible spectral window (650–850 nm) is another limitation in addition to the aggre- to the aggre-
biocompatible spectral window (650–850 nm) is another limitation in addition
gation and
gation and solubility solubility
of PSs. of PSs. SiIV phthalocyanines
SiIV phthalocyanines (SiPc), characterized
(SiPc), characterized by reduced by reduced aqueous
aque-
aggregation and high 1 O quantum yield when illuminated with tissue-penetrating far-red
ous aggregation and high 1O2 quantum 2 yield when illuminated with tissue-penetrating
light, solve
far-red light, could couldthissolve this problem
problem [60,61]. [60,61]. A positively-charged
A positively-charged PtII –SiIV phthalocyanine
PtII–SiIV phthalocya-
nine complex 11 complex
(Figure115)(Figure 5) was selectively
was selectively delivereddelivered
to cancerto cancer
cells by thecells by the hyaluronic
hyaluronic acid acid (HA)
formulated nanoparticles with the mediation of
(HA) formulated nanoparticles with the mediation of the CD44 receptor [62]. The nano- the CD44 receptor [62]. The nanoparti-
particles showed clesimproved
showed improved aqueous solubility,
aqueous solubility, specific
specific uptake, uptake, photo-enhanced
photo-enhanced cytotox- cytotoxicity
(~1500-fold) and mitochondrial accumulation in
icity (~1500-fold) and mitochondrial accumulation in CD44-overexpressed breast cancerCD44-overexpressed breast cancer cells
over normal ones in red light (45 min, 660–680 nm, 5.5 ±−22.5 mW cm−2 ). Interestingly,
cells over normal ones in red light (45 min, 660–680 nm, 5.5 ± 2.5 mW cm ). Interestingly,
the nanoconjugate delivered 11 only to cancer cells, which resulted in the generation of
cytotoxic 1O2 and PtII species.
to those kept in the dark [63]. Both complexes demonstrated a potential to serve as DNA-
targeting PDT agents. The PtII moieties lead the PDT moiety to approach DNA and execute
red-light-induced oxidative damage, while the photoactive SiPc moiety endows the PtII
units with a red-light-induced photochemical property that may lead to enhanced DNA
Pharmaceuticals 2021, 14, 133
platination. This design was expected to be superior over solo therapeutic modalities6 of and
23
obtain drugs with improved activity and reduced side effects. The use of phthalocyanine
could alleviate some limitations of PDT, however, the self-aggregation in aqueous media
may affect its photosensitivity. In brief, the Pt–porphyrin or Pt–phthalocyanine conjugates
the nanoconjugate
maintained delivered
the intrinsic 11 only
properties to cancer
of an cells,unit
individual which resulted
in cancer inand
cells, the thus
generation of
could act
cytotoxic 1 O and PtII species.
as dual threat2 anticancer agents.

Figure 5. Chemical structures of complexes 10–13.


10‒13.

Complexes
PSs based on and 13 (Figure 5)dyes
12non-macrocyclic presented a 25-used
were also and to
7-fold
design enhancement,
conjugates forrespectively,
the com-
in cytotoxicity
binative against
effect of PDT andHeLa cells at 1ofµM
inhibition DNAunder illumination
transcription. with red light
Complexes 14 andin 15
comparison
(Figure 6)
to those remarkable
showed kept in the dark [63]. Both complexes
photocytotoxicity in visibledemonstrated
light (400−700anm, potential to−2serve
10 J cm ) to theasHaCaT
DNA-
targeting PDT agents. II
and MCF-7 cells, withThe
the Pt
IC50moieties
being inlead the PDT moiety
the nanomolar level,towhile
approach
they DNA and execute
were almost non-
red-light-induced
toxic (IC50 > 80 μM) oxidative damage,
in the dark [64]. while
Complexthe photoactive
14 was emissiveSiPc and
moiety endows
showed the PtII
significant
units with a in
localization red-light-induced
the mitochondria photochemical property that
and minor localization in may lead to enhanced
the endoplasmic DNA
reticulum
platination. This design was expected to be superior over solo therapeutic
(ER), and hence could be used for cellular imaging and reducing the drawbacks associated modalities and
obtain drugs with improved activity and reduced side effects. The use of phthalocyanine
could alleviate some limitations of PDT, however, the self-aggregation in aqueous media
may affect its photosensitivity. In brief, the Pt–porphyrin or Pt–phthalocyanine conjugates
maintained the intrinsic properties of an individual unit in cancer cells, and thus could act
as dual threat anticancer agents.
PSs based on non-macrocyclic dyes were also used to design conjugates for the
combinative effect of PDT and inhibition of DNA transcription. Complexes 14 and 15
(Figure 6) showed remarkable photocytotoxicity in visible light (400−700 nm, 10 J cm−2 ) to
the HaCaT and MCF-7 cells, with the IC50 being in the nanomolar level, while they were
almost nontoxic (IC50 > 80 µM) in the dark [64]. Complex 14 was emissive and showed
Pharmaceuticals 2021, 14, 133 7 of 23

Figure 6. Chemical structures of complexes 14 and 15.


Pharmaceuticals 2021, 14, x FOR PEER REVIEW 7 of 24
Cationicsignificant
PS is helpful localization
to promote in the cellular
mitochondria and minor
accumulation localization
of anticancer in the
agents and endoplasmic
damage the reticulum
cell membrane (ER), by and hence could be
photo-induced ROSused in for
situ,cellular
which imaging
is considered and reducing
as the main the drawbacks
mechanism associated
to enhance
with with
bifunctional bifunctional
the cellular binding ofbinding
uptake ofDNA
[65]. Complex
nuclear nuclear 16DNA
(nDNA) (nDNA)
(Figure
by Pt7)drugs, by Ptsuch
exhibited drugs, such as nuclear
distinct
as nuclear excision
excision
anticancer cytotoxicity repair
repair (NER). (NER).
against MCF-7, SGC-7901, A549, and HeLa cell lines via short time
photoirradiation (532 nm, 3.5 mW cm−2, 5 min) [66]. It first accumulated on the surface of
the cell membrane in the dark for its membrane-anchoring ability, and then acted as a PS,
promoting the damage to the cell membrane in situ to increase its accumulation in tumor
cells. Although the molecular mechanism was not studied, short time photoirradiation
seems to play a key role in activating the complex.
Lysosomes as special organelles are responsible for degrading and recycling extra-
cellular materials via endocytosis and phagocytosis, and intracellular poisonous species
via autophagy [67,68]. Growing evidence indicates that lysosomes are capable of isolating
some Pt complexes
Figure toChemical
Figure6.6. reduce their
Chemical contact
structures
structures with
ofofcomplexes
complexesnDNA, 14 thus
14and
and reducing the DNA-damaging
15.
15.
effect [69]. Therefore, silencing Pt complexes in lysosomes and then activating them spe-
Cationic
cifically in the tumor tissue
Cationic PS isishelpful
PSmight be a to
helpful topromote
method
promote forthe
thecellular
cellularaccumulation
improving the antitumorof
accumulation ofanticancer
activity and agents
anticancer agentsand
and
damage
alleviating side the
effects.
damage cell
cellmembrane
theComplex membrane by
17 (Figurebyphoto-induced
7) is the firstROS
photo-induced ROSininsitu,
example of which
situ, whichisisconsidered
photoactive mono- asasthe
considered themain
main
functional Ptmechanism
II complex capable
mechanism to
toenhance
enhance the
thecellular
of lysosomal cellular uptake
escapeuptake [65].
[70]. It was
[65]. Complex
sequestered
Complex 16(Figure
16 7)7)exhibited
in lysosomes
(Figure exhibiteddistinct
distinct
anticancer
via endocytosisanticancer cytotoxicity against MCF-7, SGC-7901, A549, and HeLa cell lines via shorttime
and cytotoxicity
showed low against
cytotoxicity MCF-7,
to both SGC-7901,
normal A549,
and tumor and HeLa
cells cell
without lines
pho-via short time
photoirradiation −2
Interestingly, it(532
toirradiation.photoirradiation (532nm,
escaped nm,3.5 3.5mW
from mWthecm cm−2,, 55 min)
lysosomes min) to[66]. ItIt first
the nucleus
[66]. firstaccumulated
upon short-time
accumulated on
onthe
thesurface
surfaceofof
the
thecell
photoirradiation (532
cell membrane
nm, 3.5 mW
membrane in
inthe
cm
the−2dark for
, 5 min)
dark foritsitsmembrane-anchoring
due to the photoinduced ability,
membrane-anchoring ability and
ability,to then
thenacted
generate
and actedasasaaPS,
PS,
promoting
ROS. Apart from damaging
promoting the damage
lysosomes
the damage to the
to to cell
therelease membrane in situ
17 into theincytosol
cell membrane to
situ to andincrease its
nucleus,
increase accumulation
ROS also
its accumulation in
intumor
tumor
cells.
cells. Although
decreased intracellularAlthough GSHthe levels
the molecular
to impede
molecular mechanism was
wasnot
its deactivation
mechanism instudied,
not short
the cytosol
studied, short time
and photoirradiation
further
time photoirradiation
increased itsseems to play
accessibility a
to key
nDNA role in activating
favorable
seems to play a key role in activating the complex. for the
the complex.
antitumor activity.
Lysosomes as special organelles are responsible for degrading and recycling extra-
cellular materials via endocytosis and phagocytosis, and intracellular poisonous species
via autophagy [67,68]. Growing evidence indicates that lysosomes are capable of isolating
some Pt complexes to reduce their contact with nDNA, thus reducing the DNA-damaging
effect [69]. Therefore, silencing Pt complexes in lysosomes and then activating them spe-
cifically in the tumor tissue might be a method for improving the antitumor activity and
alleviating side effects. Complex 17 (Figure 7) is the first example of photoactive mono-
functional PtII complex capable of lysosomal escape [70]. It was sequestered in lysosomes
via endocytosis and showed low cytotoxicity to both normal and tumor cells without pho-
toirradiation. Interestingly, it escaped from the lysosomes to the nucleus upon short-time
photoirradiation
Figure 7. Chemical structures
Figure (532 nm,
of complexes
7. Chemical 3.5 mW
16 and
structures cm−2, 5 min)
of17.
complexes 16 due to the photoinduced ability to generate
and 17.
ROS. Apart from damaging lysosomes to release 17 into the cytosol and nucleus, ROS also
decreased
Pt complexes Lysosomes intracellular
combined aswithspecial GSH
PSs levels
organelles
may exhibittoare
impede its deactivation
responsible
synergistic for degrading
effects, in theand
but most ofcytosol and further
recycling
them extra-
increased
cellular its
materials accessibility
via to
endocytosisnDNA and favorable for
phagocytosis,
are limited to Pt–porphyrin conjugates. Replacing porphyrin with other PSs is another the antitumor
and activity.
intracellular poisonous species
way to developvia autophagy
effective PDT [67,68].
agents.Growing
A series evidence indicates that
of DNA-binding lysosomes are capable
PtII–triphenylamine com- of isolating
plexes were some Pt complexes
developed as potential to reduce their contact
PDT anticancer with[71–74].
agents nDNA, The thusfluorogens
reducing the DNA-damaging
π-con-
effect [69]. Therefore,
jugated triphenylamines, owing tosilencing
two-photon Pt complexes
absorption inand
lysosomes
aggregation and then activating
induced emis- them specif-
ically in the
sion (AIE) properties, aretumor
used as tissue might probes
fluorescent be a method for improving
or theranostic the antitumor
agents [75]. In order toactivity and
alleviating side effects. Complex 17 (Figure 7)
improve the PDT efficiency of the conjugates and systematically investigate the anticancer is the first example of photoactive mono-
functional PtII complex capable of lysosomal escape [70]. It was sequestered in lysosomes
via endocytosis and showed low cytotoxicity to both normal and tumor cells without pho-
toirradiation. Interestingly, it escaped from the lysosomes to the nucleus upon short-time
photoirradiation (532 nm, 3.5 mW cm−2 , 5 min) due to the photoinduced ability to generate
ROS. Apart from damaging lysosomes to release 17 into the cytosol and nucleus, ROS also
decreased intracellular GSH levels to impede its deactivation in the cytosol and further
Figure 7. Chemical structures of complexes 16 and 17.
increased its accessibility to nDNA favorable for the antitumor activity.
Pt complexes combined with PSs may exhibit synergistic effects, but most of them are
Pt complexes combined with PSs may exhibit synergistic effects, but most of them
limited to Pt–porphyrin conjugates. Replacing porphyrin with other PSs is another way to
are limited to Pt–porphyrin conjugates. Replacing porphyrin with other PSs is another
develop effective PDT agents. A series of DNA-binding PtII –triphenylamine complexes
way to develop effective PDT agents. A series of DNA-binding PtII–triphenylamine com-
were developed as potential PDT anticancer agents [71–74]. The fluorogens π-conjugated
plexes were developed as potential PDT anticancer agents [71–74]. The fluorogens π-con-
triphenylamines, owing to two-photon absorption and aggregation induced emission
jugated triphenylamines, owing to two-photon absorption and aggregation induced emis-
sion (AIE) properties, are used as fluorescent probes or theranostic agents [75]. In order to
improve the PDT efficiency of the conjugates and systematically investigate the anticancer
Pharmaceuticals 2021, 14, 133 8 of 23

maceuticals 2021, 14, x FOR PEER REVIEW 8 of 24

(AIE) properties, are used as fluorescent probes or theranostic agents [75]. In order to
improve the PDT efficiency of the conjugates and systematically investigate the anti-
cancer
SAR, trinuclear SAR, trinuclear Pt–triphenylamine
Pt–triphenylamine isomers 18−21 (Figure isomers 18−developed
8) were 21 (Figure 8) were
[76]. developed [76].
Com-
plexes 18 andComplexes
19 exhibited18 and
much exhibited
19 better PDTmuch better
activity PDT
than activity than
complexes 20 andcomplexes
21 owing 20toand 21 owing
the redder absorption and emission wavelength, higher cellular uptake and O2 quantum 1 O2 quan-
to the redder absorption and emission wavelength, higher cellular
1 uptake and
tumDNA-binding
yield, stronger yield, stronger DNA-binding
and photo-cleavage andability.
photo-cleavage ability.
In addition, In addition,
complexes 18 and complexes 18
and 19 mainly
19 mainly accumulated accumulated
in the in thecomplexes
nucleus, while nucleus, while complexes
20 and 20 and mainly
21 distributed 21 distributed
in mainly
in the
the cytoplasm. cytoplasm.
Particularly, Particularly,
complex complex
19 elicited elicitedresponses,
DNA19damage DNA damage responses,
arrested the cellarrested the
cell cycle
cycle in the G2/M in the
phase, andG2/M
led to phase, andinled
apoptosis to apoptosis
cancer cells uponin cancer cells uponatlight
light irradiation 425 irradiation
at 425 nm (40 mW cm −2 , 15 min). Moreover, it exhibited significant PDT effect on HeLa
nm (40 mW cm , 15 min). Moreover, it exhibited significant PDT effect on HeLa xenograft-
−2

bearing mice,xenograft-bearing
including reductionmice,inincluding
the tumorreduction
volume andin the tumor
cell deathvolume
in tumor andsections,
cell death in tumor
sections, but showed no noticeable side effects
but showed no noticeable side effects on body weight and major organs.on body weight and major organs.

Figure 8. Chemical structures


Figure 8.of complexes
Chemical 18‒21. of complexes 18–21.
structures

4. Targeted Monofunctional PtII Complexes


4. Targeted Monofunctional PtII Complexes
Although Pt-based drugs play an important part in cancer therapeutic regimens,
their widespreadAlthough
use is still Pt-based
limited bydrugs play toxic
the severe an important part
side effects in cancer
arising from thetherapeutic
lack of regimens,
their widespread use is still limited by the severe toxic side effects
selectivity for cancer cells. To overcome this defect, cancer-targeted Pt complexes are de- arising from the lack
of selectivity for cancer cells. To overcome this defect, cancer-targeted
veloped. The targeting group(s) in such complexes could direct the Pt warheads to cancer Pt complexes are
developed. The targeting group(s) in such complexes could direct
cells by interacting with the receptors overexpressed on the cell surface [77], or direct to the Pt warheads to cancer
the tumor ascells by interacting
a whole through the with the receptors
interaction overexpressed
with the tumor-related on cell
the cell surface
surface [77], or direct to
markers
the tumor as a whole through the interaction with the tumor-related
such as antigens or receptors [78]. Targeting could also be achieved at the subcellular level, cell surface markers
such as antigens or receptors [78]. Targeting could also be achieved
allowing Pt to be directed to specific organelles to elicit biological effects [6,78]. Herein, at the subcellular level,
allowing Pt to be directed to specific organelles to elicit biological effects [6,78]. Herein, we
we particularly focus on monofunctional PtII complexes designed for these purposes.
particularly focus on monofunctional PtII complexes designed for these purposes.
Taking osteosarcoma (OS) as an example, which is a primary malignant bone tumor
Taking osteosarcoma (OS) as an example, which is a primary malignant bone tumor
severely threatens the life of adolescents [79]. Owing to the peculiar tumor sites (knee joint
severely threatens the life of adolescents [79]. Owing to the peculiar tumor sites (knee joint
and appendicular skeleton) and lack of knowledge about driving oncogenes, as well as
and appendicular skeleton) and lack of knowledge about driving oncogenes, as well as
insufficient drug concentration in the tumor site, OS is extremely difficult to treat [80,81].
Cisplatin is ineffective for OS due to its poor accessibility and severe systemic toxicity [82].
The coordination of phosphonate groups with Ca2+ ions endows bisphosphonates with a
Pharmaceuticals 2021, 14, 133 9 of 23

Pharmaceuticals 2021, 14, x FOR PEER REVIEW 9 of 24


insufficient drug concentration in the tumor site, OS is extremely difficult to treat [80,81].
Cisplatin is ineffective for OS due to its poor accessibility and severe systemic toxicity [82].
The coordination of phosphonate groups with Ca2+ ions endows bisphosphonates with a
special affinity
special affinity for
for hydroxyapatite
hydroxyapatite in in the
the bone
bone matrix
matrix [83].
[83]. Complexes
Complexes 22 22 and
and 23
23 (Figure
(Figure 9)9)
bearing aabone-targeting
bearing bone-targetingbisphosphonate
bisphosphonate moiety
moiety exhibited
exhibited potential
potential selectivity
selectivity for OSfor[84].
OS
[84].cytotoxicity
The The cytotoxicity of cisplatin-derived
of cisplatin-derived 22 was
22 was higher
higher thanthan
thatthat of oxaliplatin-derived
of oxaliplatin-derived 23
23 towards the U2-OS cells. Bisphosphonate also improved the
towards the U2-OS cells. Bisphosphonate also improved the lipophilicity and lipophilicity and cellular
cellular
uptake of thethe complexes.
complexes. Lipophilic
Lipophilic 24 (Figure 9) was optimized to maintain the bone-
targeting property as as well
well asastotominimize
minimizethe thereactivity
reactivityofof
thethe II center
PtIIPtcenter in order
in orderto de-
to
crease the
decrease systemic
the systemictoxicity
toxicity[85].
[85].Unlike
Unlikecomplexes
complexes22 22and
and 23,
23, the
the molecular mechanism
mechanism
of complex 24 involves
involves both
both DNADNAbinding
bindingandandmevalonate
mevalonatepathway.
pathway.ItsItsacute
acutetoxicity
toxicityis
7-fold
is lower
7-fold lower than that
than of cisplatin.
that TheThe
of cisplatin. introduction of bisphosphonate
introduction of bisphosphonate provides a new
provides a
new possibility
possibility to overcome
to overcome the ineffectiveness
the ineffectiveness and systemic
and systemic toxicity
toxicity of Pt drugs
of Pt drugs for thefor the
treat-
treatment
ment of OS. of OS.

9. Chemical structures of
Figure 9. of complexes
complexes 22–24.
22‒24.

Targeting
Targetingthe thewhole
wholetumor
tumoris is
based
basedonon
thethespecific expression
specific of some
expression receptors
of some or anti-
receptors or
gens on the surface of tumor cells. Tumor-associated receptors are well
antigens on the surface of tumor cells. Tumor-associated receptors are well documented in documented in the
literature, for example,
the literature, transferrin,
for example, selectins,
transferrin, integrins,
selectins, folatefolate
integrins, receptor, glucose
receptor, transporter
glucose trans-
(GLUT), galectins, hyaluronic acid receptors, and the asialoglycoprotein
porter (GLUT), galectins, hyaluronic acid receptors, and the asialoglycoprotein receptor receptor [6,86].
Targeting these receptors
[6,86]. Targeting could selectively
these receptors deliverdeliver
could selectively a cytotoxic agent agent
a cytotoxic to cancer cells. cells.
to cancer Inte-
grins are heterodimeric
Integrins are heterodimeric transmembrane
transmembrane cell adhesion glycoproteins,
cell adhesion glycoproteins,which play play
which a keya role
key
in enhancing migration, invasion, and proliferation of cancer cells, and
role in enhancing migration, invasion, and proliferation of cancer cells, and even are linked even are linked to
tumor angiogenesis [87]. The synthesis and biological profile of a Pt II -c(RGDyK) conjugate
to tumor angiogenesis [87]. The synthesis and biological profile of a Pt -c(RGDyK) conjugate
II

25 (Figure
25 (Figure 10)
10) for
for integrin-targeted
integrin-targeted PDT PDT has
has been
been reported.
reported. Complex
Complex 25 25 was
was moderately
moderately
cytotoxic towards six cancer cell lines with different levels of integrin
cytotoxic towards six cancer cell lines with different levels of integrin expression expression [88].[88].
It wasIt
was taken up rapidly by receptor-mediated endocytosis and generated 1 O efficiently upon
taken up rapidly by receptor-mediated endocytosis and generated 1O2 efficiently 2 upon irra-
irradiation, thus showing enhanced anticancer activity as a targeted PDT agent.
diation, thus showing enhanced anticancer activity as a targeted PDT agent.
Angiogenesis is an important process required for the development of new blood
Angiogenesis is an important process required for the development of new blood ves-
vessels, and is also crucial for tumorigenesis, tumor growth, survival, and metastasis. In the
sels, and is also crucial for tumorigenesis, tumor growth, survival, and metastasis. In the
case of tumor-induced angiogenesis, transmembrane receptors such as integrins (αv β3 and
case of tumor-induced angiogenesis, transmembrane receptors such as integrins (αvβ3 and
αv β5 ) are highly expressed, which have a very high affinity for peptides containing RGD
αvβ5) are highly expressed, which have a very high affinity for peptides containing RGD
(Arg-Gly-Asp) and NGR (Asn-Gly-Arg) sequences. In this regard, complex 26 (Figure 10)
(Arg-Gly-Asp) and NGR (Asn-Gly-Arg) sequences. In this regard, complex 26 (Figure 10)
with dual antiangiogenic and antitumor activity was a non-cytotoxic compound with
with dual antiangiogenic and antitumor activity was a non-cytotoxic compound with IC50
IC50 >100 µM in different cancer cell lines (± αv β3 and αv β5 integrin receptors), while
>100 μM in different cancer cell lines (± αvβ3 and αvβ5 integrin receptors), while showing
showing the antiangiogenic activity in HUVECs at sub-cytotoxic concentrations [89], which
the antiangiogenic activity in HUVECs at sub-cytotoxic concentrations [89], which exem-
plified the design of angiogenesis inhibitors through conjugating a metallodrug with anti-
angiogenic activity to a cyclic RGD-containing peptide or a peptidomimetic analogue.
Pharmaceuticals 2021, 14, 133 10 of 23

Pharmaceuticals 2021,
Pharmaceuticals 2021, 14,
14, xx FOR
FOR PEER
PEER REVIEW
REVIEW 10 of 24
the design of angiogenesis inhibitors through conjugating a metallodrug10with
exemplified of 24
antiangiogenic activity to a cyclic RGD-containing peptide or a peptidomimetic analogue.

Figure 10.
Figure 10. Chemical
10. Chemical structures
structures of
of complexes
of complexes 25
complexes 25 and
25 and26.
and 26.
26.

Targeting angiogenesis
Targeting
Targeting provides
angiogenesis provides
providesan an alternative
analternative direction
alternativedirection
direction forfor
for tumor-targeting
tumor-targeting
tumor-targeting therapy
ther-
therapy
[90].[90].
apy
[90]. However,
However,
However, somesome
some complexes do not
complexes
complexes do not possess
dopossess aa specific
not possess specific targeting
a specific group,
targeting
targeting group, but still
group,
but still show
butshow
still
show antiangiogenic
antiangiogenic
antiangiogenic activity.
activity. activity.
For For example,
For example,
example, dinuclear
dinuclear
dinuclear complexes
complexes complexes
27–29 (Figure
27–29 (Figure
27–29 (Figure
11) were
11) were11)found
were
found
found
to to interact
to interact
interact with the
with with
the the phosphate
phosphate
phosphate backbone,
backbone,
backbone, forming
forming forming
Pt-DNA
Pt-DNA Pt-DNA
adductsadducts
adducts with
minora groove
with aa minor
with minor
groove
groove
covering covering
[91]. [91].
These These
complexes,complexes, particularly
particularly complex complex
27, are 27, are
potential
covering [91]. These complexes, particularly complex 27, are potential chemotherapeutics potential chemothera-
chemotherapeutics
peutics
with with anticancer
with anticancer
anticancer and and antiangiogenic
and antiangiogenic
antiangiogenic activities,activities,
activities, and no
and and no
no toxic
toxic toxicat
effects
effects ateffects at the desired
the desired
the desired concen-
concen-
concentration.
tration. They
tration. They
They overcame overcame
overcame cisplatin cisplatin
cisplatin resistance resistance
resistance inin the in the
the zebrafish–mousezebrafish–mouse
zebrafish–mouse melanoma melanoma
melanoma xenograft
xenograft
xenograft
model and
model andmodel and effectively
effectively
effectively blocked tumor
blocked blocked
tumor tumor neovascularization
neovascularization
neovascularization and melanoma
and melanoma and melanoma
cell metastasis.
cell cell
metastasis.
metastasis.
The activation
The activationTheofactivation
of these of these complexes
these complexes
complexes resultmay
may result
may fromresult
from their from
their their
positive
positive positive
charge
charge (+4)charge
(+4) the(+4)
at the
at at
physi-
physi-
the physiological
ological conditions conditions
and and
affinity affinity
for DNA, for DNA,
heparan heparan
sulphate sulphate
(HS),
ological conditions and affinity for DNA, heparan sulphate (HS), and enzyme heparanase and (HS),
enzyme and enzyme
heparanase
heparanase
(HPSE). It
(HPSE). is(HPSE).
It is worth It is worth
worth noting
noting thatnoting
that these that these complexes
these complexes
complexes showed no
showed noshowed
sign of
sign no sign of cardiovascu-
of cardiovascular
cardiovascular toxicity
toxicity
lar toxicity
such as such
as pericardial as pericardial
pericardial edemaedema or edema
or disturbedor disturbed
disturbed heart
heart beat heart
beat rate, beat
rate, and rate,
and liver and
liver toxicityliver
toxicity suchtoxicity
such as such
as liver as
liver ne-
ne-
such
liver necrosis,
crosis, liver
liver size liver size
size change
change or change or
or reduced reduced
reduced yolk yolk
yolk absorption,absorption,
absorption, whichwhich are which
are the are
the major the major
major obstacles obstacles
obstacles limiting
limiting
crosis,
limiting
the the long-term
long-term application application
of of anticancer
clinical clinical anticancer
drugs. drugs.
the long-term application of clinical anticancer drugs.

Figure 11.
Figure
Figure 11. Chemical
11. Chemical structures
structures of
of complexes
of complexes 27–29.
complexes 27‒29.
27‒29.

Organelle-targeting
Organelle-targeting anticancer
Organelle-targeting anticancer agents
anticancer agents add
agents add aaa new
add new dimension
new dimension to
dimension to the
to the discovery
the discovery and
discovery and de-
de-
velopment
velopment ofofPtPtdrug
drugcandidates.
candidates. Among
Among different organelles,
different mitochondria
organelles,
velopment of Pt drug candidates. Among different organelles, mitochondria have re- mitochondriahave received
have re-
much
ceived attention
much in recent
attention in years.
recent The oxidative
years. The phosphorylation
oxidative (OXPHOS)
phosphorylation
ceived much attention in recent years. The oxidative phosphorylation (OXPHOS) and gly- and
(OXPHOS) glycolysis
and gly-
in mitochondria
colysis
colysis in offer nutrients
in mitochondria
mitochondria offer and energy
offer nutrients
nutrients and to cancer
and energy
energy cells for
to cancer
to cancer occurrence,
cells
cells growth,
for occurrence,
for occurrence, and
growth,
growth,
transformation
and transformation
and [92,93].
transformation [92,93]. Nevertheless, unlike
[92,93]. Nevertheless,
Nevertheless, unlikenormal
unlike normalcells, most
normal cells, cancer
cells, most
most cancer cells preferentially
cancer cells
cells preferen-
preferen-
use aerobic
tially glycolysis
use aerobic
aerobic as theas
glycolysis metabolic pathway
the metabolic
metabolic for glucose,
pathway whichwhich
for glucose,
glucose, is accompanied
is accompanied
accompaniedby a
tially use glycolysis as the pathway for which is
high rate
by aa high of
high rate glucose
rate of consumption
of glucose
glucose consumption and
consumption andlactate production,
and lactate
lactate production,even
production, even when
even when oxygen
when oxygen is available
oxygen is is avail-
avail-
by
for
ableOXPHOS
for OXPHOS [94–96]. This This
[94–96]. abnormal energy
abnormal metabolism
energy process
metabolism involves
process many
involves proteins
many pro-
able for OXPHOS [94–96]. This abnormal energy metabolism process involves many pro-
and
teins enzymes,
and thus thus
enzymes, providing potential
providing targets
potential for for
targets thethe
design
design ofofanticancer
anticancer drugs
drugs and
and
teins and enzymes, thus providing potential targets for the design of anticancer drugs and
overcoming
overcoming the the drug
the drug resistance.
drug resistance.
resistance.
overcoming
It is
It is generally
generally believed
believed that
that nDNA
nDNA isis the
the primary
primary target
target of
of Pt-based
Pt-based anticancer
anticancer drugs,
drugs,
and the resistance to Pt agents mainly result from the extensive repair
and the resistance to Pt agents mainly result from the extensive repair of Pt-DNA adducts of Pt-DNA adducts
Pharmaceuticals 2021, 14, 133 11 of 23

Pharmaceuticals 2021, 14, x FOR PEER REVIEW 11 of 24

It is generally believed that nDNA is the primary target of Pt-based anticancer drugs,
and the resistance to Pt agents mainly result from the extensive repair of Pt-DNA adducts
bythe
by theactivation
activationofofDNA DNA repair
repair mechanisms
mechanisms in tumor
in tumor cells
cells [97–99].
[97–99]. Whereas,
Whereas, mitochon-
mitochondria
dria contain
contain their their
ownown cycliccyclic mitochondrial
mitochondrial DNA DNA (mtDNA),
(mtDNA), which
which is ismore
morevulnerable
vulnerableto to
damage than
damage than nDNA
nDNA due due totothethelack
lackofofhistone
histone protection
protection andandproximity
proximityto the ROSROS
to the pro-
duction sitesite
production [100,101].
[100,101].Therefore, mtDNA
Therefore, mtDNAis a potential target target
is a potential for potentiating the activity
for potentiating the
activity of anticancer drugs. A cationic naphthalimide-modified complex 30 (Figurenot
of anticancer drugs. A cationic naphthalimide-modified complex 30 (Figure 12) 12) only
not
caused
only severe
caused nDNA
severe nDNA damage
damage butbutalso
alsoinduced
inducedthe themtDNA
mtDNAlesion lesion and regulated the
and regulated the
downstreamgene
downstream geneexpression
expression ofof mtDNA-encoded
mtDNA-encoded proteins
proteins [102].
[102]. Moreover,
Moreover, it disturbed
it disturbed the
the physiological
physiological process process of mitochondria
of mitochondria by reducing
by reducing the mitochondrial
the mitochondrial membrane
membrane po-
potential
tential (MMP)
(MMP) and promoting
and promoting the generation
the generation of ROS. of Dinuclear
ROS. Dinuclear complex complex 31 (Figure
31 (Figure 12) was12)
was composed
composed of IrIIIofand
IrIII PtII moieties.
and PtII moieties. It accumulated
It accumulated in the
in the mitochondria
mitochondria by aby a rate
rate of to
of up up
to 76% with an energy-independent uptake mechanism [103].
76% with an energy-independent uptake mechanism [103]. The complex exhibited strong The complex exhibited
strong antitumor
antitumor activity activity
towardstowards A549R cisplatin-resistant
A549R cisplatin-resistant cancer cellscancer
and cells and damaged
damaged the mtDNA the
mtDNA severely.
severely. Further, Further,
it disrupted it disrupted the mitochondrial
the mitochondrial function,function,
resultedresulted
in a lossinofa loss
MMP, of
depleted ATP, and
MMP, depleted finally
ATP, andinduced necrosis necrosis
finally induced to cancertocells.
cancerAllcells.
theseAll
findings suggest that
these findings sug-
mtDNA-targeted
gest that mtDNA-targetedPt complexes are potential
Pt complexes are antitumor agents against
potential antitumor agentscisplatin-resistant
against cisplatin-
cancer cells.
resistant cancer cells.

Figure12.
Figure 12.Chemical
Chemicalstructures
structuresof
ofcomplexes
complexes30
30and
and31.
31.

Recently,we
Recently, weinvestigated
investigatedthe theanticancer
anticancermechanism
mechanismof ofthree
threemitochondrion-targeted
mitochondrion-targeted
PtIIII complexes
Pt complexes32 32−34 (Figure13)
−34 (Figure 13)from
fromthethe perspective
perspective of DNA
of DNA damage,
damage, energy
energy metabo-
metabolism,
lism,SAR
and and [104].
SAR [104].
AmongAmong them,them, complex
complex 32 exhibited
32 exhibited greater
greater inhibitoryactivity
inhibitory activityononthe
the
A549cells
A549 cellsthan
than cisplatin
cisplatin in vitro and vivo. Moreover,
Moreover,ititnot
notonly
onlycombined
combinedwith withnDNA
nDNA in
a monodentate
in a monodentate manner
manner andanddamaged
damaged mtDNA,
mtDNA, but but
alsoalso
inhibited glycolysis
inhibited of cancer
glycolysis cells,
of cancer
cells,
affected affected the structure
the structure and and function
function of mitochondria,
of mitochondria, resulting
resulting in an
in an abnormal
abnormal process
process of
of mitochondrial
mitochondrial OXPHOS
OXPHOS andand tricarboxylic
tricarboxylic acidacid cycle.
cycle. In vivo
In vivo studies
studies showed
showed thatthat as
as the
the
tumor tumor shrank,
shrank, thethe body
body weight
weight of 32-treated
of 32-treated mice
mice alsoalso reduced,
reduced, which
which may may be due
be due to
to the
the mitochondrion-disrupting
mitochondrion-disrupting effect.
effect. Moreover,
Moreover, mostmost of the
of the Pt accumulated
Pt accumulated in the
in the liverliver
and
and
kidneys,kidneys, implying
implying that that the cellular
the cellular uptakeuptake
of 32of 32 may
may be mediated
be mediated by organic
by organic cationcation
trans-
Pharmaceuticals 2021, 14, x FOR PEERtransporters,
REVIEW which are primarily expressed in these organs. This study provides 12new
of 24
porters, which are primarily expressed in these organs. This study provides new insights
insights
into the into the mechanism
mechanism of actionoffor
action for Pt anticancer
Pt anticancer drugs. drugs.
Another mitochondrion-targeted complex 35 (Figure 13) that modified by tri-
phenylphosphonium can modulate signaling pathways relevant to cancer bioenergetics
[105]. It enhanced cytotoxicity against cisplatin-insensitive Caov-3 cells, exerted inhibition
to mitochondrial thioredoxin reductase (TrxR), damaged mitochondrial morphology and
function, destroyed both respiratory and glycolytic metabolisms, and induced cancer cells
to enter into a hypometabolic state. The results highlight that targeting redox homeostasis
and modulating metabolic pathways could effectively improve the anticancer effect.

Figure 13.
Figure 13. Chemical
Chemical structures
structures of
of complexes
complexes 32–35.
32‒35.

In order to overcome the shortcomings of classic Pt drugs, extensive research has


been initiated to search for new targets other than DNA. Enzymes play vital roles in al-
most all physiological and pathophysiological processes, and have long been considered
as drug targets [106]. It is estimated that more than 47% of drugs target enzymes [107].
Pharmaceuticals 2021, 14, 133 12 of 23

Another mitochondrion-targeted complex 35 (Figure 13) that modified by triph-


enylphosphonium can modulate signaling pathways relevant to cancer bioenergetics [105].
It enhanced cytotoxicity against cisplatin-insensitive Caov-3 cells, exerted inhibition to
mitochondrial thioredoxin reductase (TrxR), damaged mitochondrial morphology and
function, destroyed both respiratory and glycolytic metabolisms, and induced cancer cells
to enter into a hypometabolic state. The results highlight that targeting redox homeostasis
and modulating metabolic pathways could effectively improve the anticancer effect.
In order to overcome the shortcomings of classic Pt drugs, extensive research has been
initiated to search for new targets other than DNA. Enzymes play vital roles in almost all
physiological and pathophysiological processes, and have long been considered as drug
targets [106]. It is estimated that more than 47% of drugs target enzymes [107]. Therefore,
enzyme inhibition could be a significant and alternative mechanism for Pt-based anticancer
drugs. Protein tyrosine phosphatases (PTPs), a superfamily of enzymes, participate in the
regulation of the intracellular signal transduction pathway by removing the phosphate
groups from proteins [108]. Dysregulated activities of PTPs are related to the pathogenesis
of many human diseases such as cancers, diabetes, and autoimmune diseases [108,109].
Complex 36 (Figure 14) displayed an antiproliferative activity against MCF-7 cells superior
to cisplatin [110]. It selectively inhibited PTP1B, thus significantly influenced the cellular
phosphorylation level and further the intracellular signal transduction pathway, which is
Pharmaceuticals 2021, 14, x FOR PEERdistinctly
REVIEW different from the DNA-damaging mechanism for cisplatin, thereby providing 13 of 24 a
new clue for designing Pt-based anticancer drugs.

Figure 14.
Figure 14. Chemical
Chemicalstructures
structuresofofcomplexes
complexes36‒39.
36–39.

Telomerase
Hexokinaseisispresent
the first in the majority (85–90%)
rate-limiting enzymeofincancer cells but is
the glycolytic undetectable
pathway, in
catalyzing
normal
the cell lines,ofwhich
production is restricted by thefrom
glucose-6-phosphate levelglucose
of hTERT and In
[111]. c-myc proteins
normal cells,[114,115].
hexokinase
Complexeshave
isozymes 40−41low(Figure 15) inducedexpression
transcriptional apoptosis in the NCI-H460
levels and each cells via inhibiting
of them has tissuethe speci-
telomerase
ficity, whileandas adisrupting
key enzyme the of
function of mitochondria
glycolysis, hexokinaseatis0.89 and 0.10
widely and μM, respectively
highly expressed in
[116,117].
cancer In particular,
cells, which not41 significantly
only promotesinhibited the growth of
aerobic glycolysis, tumor
but also in NCI-H460
increases thetumor-
resistance
bearing mice with the tumor growth inhibition rate (TGI) of 40.7% and
to cell death signals [112]. Complexes 37−39 (Figure 14), anchoring lonidamine (an in- no obvious toxicity.
Complex
hibitor 42 (Figure
of hexokinase) to15)
thecontaining
PtII center,acould
jatrorrhizine derivative
selectively reducealso
the exhibited a remark-
bioenergetics of cancer
able antitumor activity and lower general toxicity in vitro and vivo compared
cells [113]. Particularly, 39 showed higher cytotoxicity than cisplatin against MDA-MB-231 to cisplatin
[118].(9.3
cells It displayed
µM), caused highsignificant
selectivity damage
for HeLatocells (IC50 =and
mtDNA, 1.00 disrupted
± 0.17 nM)mitochondrial
by targeting p53 bioen-
and telomerase, and showed green luminescence. In addition, it caused
ergetics. These complexes perturbed the signal pathways related to cell death, includingmitochondrial and
DNA damage,
DNA damage, and induced a process,
the metabolic high rateandof apoptosis evenregulatory
transcription at a low dose of 1.00 nM. The
activity.
HeLa tumor inhibition rate (TIR) of 42 (48.8%) was even higher than that of cisplatin
Telomerase is present in the majority (85–90%) of cancer cells but is undetectable in
(35.2%). The low systemic toxicity of 42 is quite impressive, in that the body weight of the
normal cell lines, which is restricted by the level of hTERT and c-myc proteins [114,115].
treated mice (mstart = 18.6 ± 0.5 g, mend = 20.1 ± 0.5 g) was hardly affected as compared with
the control group (mstart = 18.7 ± 1.2 g, mend = 20.7 ± 1.4 g). Similarly, complex 43 (Figure 15)
exerted cytotoxicity mainly via inhibiting telomerase by interaction with the c-myc quad-
ruplex and disruption of the mitochondrial function [119]. The complex exhibited selec-
tive cytotoxicity to T-24 cells.
Pharmaceuticals 2021, 14, 133 13 of 23

Complexes 40−41 (Figure 15) induced apoptosis in the NCI-H460 cells via inhibiting
the telomerase and disrupting the function of mitochondria at 0.89 and 0.10 µM, respec-
tively [116,117]. In particular, 41 significantly inhibited the growth of tumor in NCI-
Pharmaceuticals 2021, 14, x FOR PEERH460
REVIEWtumor-bearing mice with the tumor growth inhibition rate (TGI) of 40.7% and
14 of 24 no
obvious toxicity.

Figure 15.
Figure 15. Chemical
Chemicalstructures
structuresofofcomplexes
complexes40‒43.
40–43.

5. Miscellaneous
Complex 42 Monofunctinoal PtII Complexes
(Figure 15) containing a jatrorrhizine derivative also exhibited a re-
markable antitumor
Generally, Pt drugsactivity
induce and lower
cancer cellgeneral toxicity
death by in vitro
interfering withandDNAvivo compared
synthesis or to
cisplatin [118]. Itdamage
causing chemical displayed high which
to DNA, selectivity for HeLa
is mainly cells (IC
manifested = 1.00 ±However,
by50apoptosis. 0.17 nM) by
targeting
accumulatingp53 evidences
and telomerase,
indicateandthat showed
Pt drugs green luminescence.
may have other molecularIn addition,
targets in it caused
addi-
mitochondrial
tion to DNA, which and DNA damage,
can induce and induced
cell death through anon-apoptotic
high rate of pathways,
apoptosis such
evenasatau-
a low
dose of 1.00
tophagy, nM. The
necrosis, andHeLa
even tumor inhibition [105,120–123].
immunogenicity rate (TIR) of 42 (48.8%) was
Complexes even
44–46 higher
(Figure 16)than
that of cisplatin
showed (35.2%). The
a dose-dependent low systemicactivity
antiproliferative toxicity
in of
the42 is quite
A2780 impressive,
cells, in that the
with the cytotoxi-
city order
body of 44of< the
weight 45 <treated
46, by amice
combinative 18.6 ± 0.5
(mstart =apoptotic mechanism = 20.1 ± 0.5
g, mend involving mitochondrial
g) was hardly
and autophagic
affected as comparedpathways
with[124]. Complex
the control 47 (Figure
group (mstart = not±only
16)18.7 initiated
1.2 g, mend =a20.7 ± 1.4
series of g).
events associated
Similarly, complex with mitochondrial
43 (Figure dysfunction,
15) exerted but also
cytotoxicity induced
mainly an apparenttelomerase
via inhibiting ER stress by
through thewith
interaction ROSthe release
c-mycand TrxR inhibition
quadruplex [125]. It of
and disruption simultaneously caused
the mitochondrial intrinsic
function [119].
pathway-dependent
The complex exhibited apoptosis
selective and apoptosis-dependent
cytotoxicity to T-24 cells.pro-death autophagy in A549
cells. The interactions of 48 (Figure 16) with different topologies of DNA imply that it
interacted
5. with DNA
Miscellaneous non-covalently,Pt
Monofunctinoal II Complexes
but could degrade once reacted with proteins, form-
ing adducts with different Pt/protein ratios [126]. This unusual mechanism of action may
Generally, Pt drugs induce cancer cell death by interfering with DNA synthesis or
origin from the peculiar reactivity with biomacromolecules.
causing chemical damage to DNA, which is mainly manifested by apoptosis. However,
accumulating evidences indicate that Pt drugs may have other molecular targets in addition
to DNA, which can induce cell death through non-apoptotic pathways, such as autophagy,
necrosis, and even immunogenicity [105,120–123]. Complexes 44–46 (Figure 16) showed
a dose-dependent antiproliferative activity in the A2780 cells, with the cytotoxicity order
of 44 < 45 < 46, by a combinative apoptotic mechanism involving mitochondrial and
autophagic pathways [124]. Complex 47 (Figure 16) not only initiated a series of events
associated with mitochondrial dysfunction, but also induced an apparent ER stress through
the ROS release and TrxR inhibition [125]. It simultaneously caused intrinsic pathway-
dependent apoptosis and apoptosis-dependent pro-death autophagy in A549 cells. The
interactions of 48 (Figure 16) with different topologies of DNA imply that it interacted with
DNA non-covalently, but could degrade once reacted with proteins, forming adducts with
different Pt/protein ratios [126]. This unusual mechanism of action may origin from the
peculiar reactivity with biomacromolecules.

Figure 16. Chemical structures of complexes 44‒48.


events associated with mitochondrial dysfunction, but also induced an apparent ER stress
through the ROS release and TrxR inhibition [125]. It simultaneously caused intrinsic
pathway-dependent apoptosis and apoptosis-dependent pro-death autophagy in A549
cells. The interactions of 48 (Figure 16) with different topologies of DNA imply that it
Pharmaceuticals 2021, 14, 133 interacted with DNA non-covalently, but could degrade once reacted with proteins, form-
14 of 23
ing adducts with different Pt/protein ratios [126]. This unusual mechanism of action may
origin from the peculiar reactivity with biomacromolecules.

Figure
Pharmaceuticals 2021, 14, x FOR PEERFigure
REVIEW16.
16. Chemical
Chemicalstructures
structuresofofcomplexes
complexes44‒48.
44–48. 15 of 24

Some monofunctional PtII complexes exhibit unique properties due to special struc-
tures.Some
Complex 49 (FigurePt17)
monofunctional could weaken
II complexes exhibit the viability
unique and invasibility
properties due to special ofstruc-
the human
seminoma cells through the PI3K/Akt signaling and mitochondria-mediated
tures. Complex 49 (Figure 17) could weaken the viability and invasibility of the human apoptotic
pathways
seminoma [127]. It may the
cells through serve as a potential
PI3K/Akt signaling drug
andin the treatment of testicular
mitochondria-mediated germ cell
apoptotic
tumors.
pathwaysComplexes
[127]. It may 50− 53 (Figure
serve 17) showed
as a potential drug indifferent biological
the treatment activitiesgerm
of testicular owingcellto their
different conformations,
tumors. Complexes among 17)
50−53 (Figure which 50 and
showed 51 with
different a cis configuration
biological exhibited
activities owing to their higher
anticancer activity thanamong
different conformations, 52 and 53 with
which 50 anda trans
51 withconfiguration towards
a cis configuration cancer
exhibited cells [128].
higher
Concretely, complexes
anticancer activity than 52 and5351with
50and showed a high
a trans affinity for
configuration the minor
towards cancergrooves of DNA,
cells [128].
Concretely,
while 52 and complexes 50 and bind
53 moderately 51 showed
to theamajor
high affinity
groovesforofthe
DNA. minorThegrooves
enhancedof DNA,
anticancer
while 52 of
activity and 5053and
moderately
51 may be bind to the major
attributed grooves
to their of DNA.
higher The enhanced
affinity for nDNA anticancer
due to the for-
activity of 50 and 51 may be attributed to their higher affinity
mation of aqua species in the cell culture. All these complexes significantlyfor nDNA due to the for-
increased
mation of aqua species in the cell culture. All these complexes significantly
the generation of ROS, which consequently depolarized the mitochondrial membrane increased the
generation of ROS, which consequently depolarized the mitochondrial
and damaged the nDNA. Thus, the cis-complexes can be regarded as mitochondrial and membrane and
damaged the nDNA. Thus, the cis-complexes can be regarded as mitochondrial and DNA-
DNA-targeting anticancer agents. In complex 54 (Figure 17), ferrocenyl terpyridine led
targeting anticancer agents. In complex 54 (Figure 17), ferrocenyl terpyridine led to a dra-
to a dramatic decrease in the dark toxicity [129]. However, it showed a low lying broad
matic decrease in the dark toxicity [129]. However, it showed a low lying broad absorption
absorption band at 600 nm and excellent ROS-mediated photocytotoxicity in visible light,
band at 600 nm and excellent ROS-mediated photocytotoxicity in visible light, with IC50
with
valuesICof values
50 9.5 and 12of μM
9.5 and 12 µMand
in HaCaT in HaCaT andlines,
MCF-7 cell MCF-7 cell lines, which
respectively, respectively, which
imply that 54 imply
that could
could54act as anact as an photoinitiator
photoinitiator in visible light.
in visible light.

Figure 17.
Figure 17.Chemical
Chemicalstructures of of
structures complexes 49‒54.
complexes 49–54.

Considering
Consideringthe thetimeliness
timelinessandandnovelty of the
novelty of literature, the above
the literature, mentioned
the above com- com-
mentioned
pounds do
pounds do not
notcover
coverallallthe
themonofunctional
monofunctional PtII Pt II complexes
complexes and their research
and their scope.scope.
research If If
readers are
readers areinterested
interestedininmore
more details on on
details thethe
above complexes,
above please
complexes, refer to
please the to
refer original
the original
papers and
papers andthe
thefollowing
followingsummary
summary Table 1. 1.
Table
Pharmaceuticals 2021, 14, 133 15 of 23

Table 1. Summary of monofunctional PtII complexes 1−54.

Complex Functional Group Function Tested Cells or Animals Ref.


Fluorescent Complexes
1 anthraquinone monitor subcellular localization U2-OS, U2-OS/Pt, A2780, A2780/DDP [40]
2, 3 4-nitrobenzo-2-oxa-1,3-diazole track cellular distribution HeLa [43]
4 4-amino-7-nitro-2,1,3- benzoxadiazole in vitro and vivo fluorescence imaging MCF-7, A549, 293T; zebrafish larva [44]
5 ThT derivative cellular imaging HeLa [45]
Photoactive Complexes
6, 7 isomeric tetra-cationic(pyridyl)porphyrins PDT on metastatic melanoma cells WM1366 [56]
8 5,10,15,20-tetra-(4-pyridyl)-21H,23H-porphine photocytotoxicity (50 W LED light, 6 J cm−2 , 30 min) colon26, sarcoma180; colon26 tumor-bearing mice [57]
photocytotoxicity (6.95 J cm−2 , 420 nm, 15 min),
9 5,10,15,20-tetra(4-pyridyl)porphyrin MRC-5, HeLa, A2780, CP70 [51]
DNA photocleavage
10 porphyrin containing GaIII center singlet oxygen generator, photocytotoxicity, DNA interaction colon 26, sarcoma 180; colon26 tumor-bearing mice [59]
specific cellular uptake, mitochondrial accumulation,
11 SiIV phthalocyanine MDA-MB-231, HEK293T [62]
photocytotoxicity (45 min, 660–680 nm, 5.5 ± 2.5 mW cm−2 )
12, 13 SiIV phthalocyanine photocytotoxicity, DNA-targeting PDT agents HeLa [63]
4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) and its photocytotoxicity (400−700 nm,
14, 15 HaCaT, MCF-7 [64]
diiodo derivative 10 J cm−2 ), cellular imaging
photocytotoxicity (532 nm,
16 α-(4-amino)styryl-4,4-difluoro-4-bora-3a,4a-diaza-s-indacene MCF-7, SGC-7901, A549, HeLa [66]
3.5 mW cm−2 , 5 min)
photocytotoxicity, lysosomal escape, increase accessibility to
17 α-(4-amino)styryl-4,4-difluoro-4-bora-3a,4a-diaza-s-indacene MCF-7, SGC-7901, A549, HeLa [70]
nDNA, decrease intracellular GSH
HeLa, HepG2, A549, A549cisR, LO2; HeLa
18–21 triphenylamine core PDT activity [76]
xenograft-bearing nude mice
Targeted Complexes
22–23 bisphosphonate bone targeting U2-OS, MG-63; male ICR mice [84]
24 bisphosphonate bone targeting, decrease systemic toxicity U2-OS, MG-63, LO2; male ICR mice [85]
25 c(RGDyK) tumor targeting, integrin-targeted PDT SKOV-3, PC-3, A549, MCF-7, MDA-MB-231, U87M [88]
26 cyclic peptide containing RGD sequence (-Arg-Gly-Asp-) target angiogenesis, antiangiogenic and antitumor activity SK-MEL-28, MDA-MB-231, CAPAN-1, HUVEC [89]
4,4’-bipyridine, 1,2-di(pyridin-4-yl)ethane, or target angiogenesis, overcome cisplatin resistance, block tumor MRC-5, A549, A375; B16-F10 melanoma-zebrafish,
27–29 [91]
1,2-di(pyridin-4-yl)ethene neovascularization and metastasis HCT-116-zebrafish
target mtDNA, damage mtDNA, regulate mtDNA-encoded
30 naphthalimide MCF-7, A549, Caov-3, HK-2; MCF-7 tumor-bearing mice [102]
protein, disturb mitochondrial physiological process
IrIII moiety plus imidazo[4,5-f][1,10]phenanthroline target mtDNA, accumulate in mitochondria, induce
31 HepG2, HeLa, A549, A549R [103]
derivative mitochondrial dysfunction via mtDNA damage
target mtDNA, inhibit glycolysis, affect mitochondrial structure
32–34 triphenylphosphonium A549, HeLa, SMMC, HL-7720; A549 tumor-bearing mice [104]
and function, damage mtDNA
target mitochondrion, inhibit mitochondrial TrxR, destroy
35 triphenylphosphonium Caov-3, A549, A549R, HK-2 [105]
respiratory and glycolytic metabolisms
target tyrosine phosphatases, selectively inhibit PTP1B,
36 5-chlorosalicylideneaniline MCF-7, HepG2, A549 [110]
antiproliferative activity
Pharmaceuticals 2021, 14, 133 16 of 23

Table 1. Cont.

Complex Functional Group Function Tested Cells or Animals Ref.


target hexokinase, disrupt mitochondrial bioenergetics,
37–39 lonidamine A549, PC3, Caov-3, MCF-7, MDA-MB-231, MCF-10A [113]
damage mtDNA
target telomerase, inhibit telomerase, disrupt SKOV-3, NCI-H460, HeLa, HL-7702, BEL-7402; NCI-H460
40, 41 naphthalene imide derivatives [116,117]
mitochondrial function tumor-bearing mice
target telomerase and p53, cause mitochondrial and DNA SKOV-3/DDP, T-24, HeLa, HL-7702, A549; HeLa
42 jatrorrhizine derivative [118]
damage, display antitumor activity and green luminescence tumor-bearing mice
target telomerase, inhibit telomerase by interacting with c-myc
43 4-([2,20 :60 ,20 0 -terpyridin]-40 -yl)-N,N-diethylaniline BEL-7404, A549, MGC80-3, T-24, HL-7702 [119]
quadruplex, disrupt mitochondrial function
Miscellaneous Complexes
9-anthryl, 9-phenantryl, and 1-pyrenyl participate in apoptotic mechanism involving mitochondrial and
44–46 MCF-7, PC3, HCT-116, A2780, Fibroblasts [124]
2,6-bis(thiazol-2-yl)pyridines autophagic pathways
BEL-7404, SKOV-3, HepG2, HCT-116, HL-7702; A549
47 8-substituted quinoline derivatives induce ER stress, cause apoptosis and pro-death autophagy [125]
tumor-bearing mice
48 terpyridine with two piperidine substituents peculiar reactivity with biological macromolecules (proteins) hen egg white lysozyme (HEWL, protein) [126]
induce apoptosis via PI3K/Akt signaling and
49 6,7-dichloro-5,8-quinolinedione TCam-2, SEM-1 [127]
mitochondria-mediated apoptotic pathways
50–53 mono- and dialkylphenylphosphines conformation-dependent biological activity MCF-7, A549, BEAS-2B, HCT-116 [128]
photocytotoxicity in visible light
54 ferrocenyl-terpyridine HaCaT [129]
(400–700 nm)
Pharmaceuticals 2021, 14, 133 17 of 23

6. Conclusions
Platinum-based anticancer agents are the mainstay of chemotherapy regimens. Their
drawbacks such as inherent or acquired drug resistance and systemic toxicity have stimu-
lated the exploration of new possible drugs. Monofunctional PtII complexes are a potential
new type of metallodrugs that break the traditional structure-activity relationships of plat-
inum drugs and exhibit improved therapeutic efficacy. In this review, we introduced the
basic conception of monofunctional PtII complexes and summed up some representative
properties and potential applications. Fluorescent monofunctional complexes have the
potentiality to monitor their distribution and travelling track in vitro and vivo with a
high temporal and spatial resolution, which would help in understanding the therapeutic
process of the complexes. Photoactive monofunctional complexes combine chemotherapy
with photodynamic therapy, which provide a dual mechanism involving light-induced
ROS and direct DNA damage to potentiate the action of PDT in hypoxic regions and
overcome the drug resistance. Targeted monofunctional complexes could increase the
amount of Pt content at the tumor site and avoid the side reactions with normal cells,
thereby enhancing the efficacy and reducing the systemic toxicity of the complexes. All
these characteristics have gone beyond the properties of existing platinum-based anticancer
drugs. Simplicity in the synthesis in comparison with organic drugs is another advantage
of monofunctional PtII complexes, or rather, all kinds of Pt complexes. Typically, there
are three synthetic routes to these complexes. (1) For [Pt(NH3 )2 (Am)Cl]+ complexes, one
chloride ion in cisplatin was first removed by silver nitrate or silver sulfate in dimethylfor-
mamide (DMF), and amine (Am) was then added to the solution. The goal product was
obtained after the addition of diethyl ether or other organic solvents. (2) For [Pt(3Am)Cl]+
(Am = aromatic N-heterocyclic amine) complexes, a direct reaction between potassium
tetrachloroplatinate(II) (K2 PtCl4 ) and Am in dimethyl sulfoxide (DMSO) would give the
target product. (3) Alternatively, cis-[Pt(DMSO)2 (Cl)2 ] or bis(benzonitrile) dichloroplat-
inum(II) [Pt(PhCN)2 Cl2 ] was mixed with Am in an organic solution (methanol or a mixture
of methanol and acetonitrile/DMSO/acetone) to obtain the monofunctional PtII complex.
If necessary, Am could be functionalized beforehand.
Last but not least, current researches on monofunctional PtII complexes are largely
limited to the molecular and cellular levels, or at best to simple animal tests. Pharma-
cokinetic and clinical trial data are completely absent, which greatly hinder any objective
assessment for the prospective development of these drug candidates. Recently, our studies
indicate that some monofunctional PtII complexes do not react with DNA but still display
anticancer activity [105,130]. The findings suggest that the known mechanism of action for
these complexes is not impeccable, and many unknown facts need to be revealed in the
future. In some less focused sides, the identification of molecular target and target-oriented
molecular design, as well as the revealing of a new anticancer mechanism would be a
meaningful aspect for the research of monofunctional PtII complexes.

Author Contributions: Conceptualization, X.W, and Z.G.; writing—original draft preparation, S.J.
and Y.G.; writing—review and editing, X.W.; supervision, X.W.; funding acquisition, X.W. and Z.G.
All authors have read and agreed to the published version of the manuscript.
Funding: We acknowledge the financial support from the National Natural Science Foundation of
China (grants 31570809, 21877059, 21731004, 91953201), the Natural Science Foundation of Jiangsu
Province (BK20202004), and the Key Scientific Research Project of Colleges and Universities in He’nan
Province (21A150009).
Conflicts of Interest: The authors declare no conflict of interest.
Pharmaceuticals 2021, 14, 133 18 of 23

Appendix A

Table A1. Designations of cell lines.

Codes Designated Cell Lines


A2780; CP70 human ovarian cancer cell
A2780/DDP cisplatin-resistant human ovarian cancer cell
A375 human melanoma cell
A549 human lung carcinoma cell
A549cisR; A549R cisplatin-resistant human lung carcinoma cell
B16-F10 mouse melanoma cell
BEAS-2B human normal bronchial epithelial cell
BEL-7402 human hepatoma cell
Caov-3 human ovarian cancer cell
CAPAN-1 human pancreatic adenocarcinoma cell
colon26 murine colon carcinoma cell
HaCaT human keratinocyte
HCT-116 human colon carcinoma cell
HEK293T; 293T human embryonic kidney cell
HeLa human cervical carcinoma cell
HepG2 human hepatocarcinoma cell
HK-2 human renal tubular epithelial cell
HL-7702; LO2 human normal liver cell
HUVEC human umbilical vein endothelial cell
MCF-10A human normal breast epithelial cell
MCF-7 human breast adenocarcinoma cell
MDA-MB-231 human breast cancer cell
MG-63 human osteosarcoma cell
MGC80-3 human gastric adenocarcinoma cell
MRC-5 human normal lung fibroblast cell
NCI-H460 human lung carcinoma cell
PC-3; PC3 human prostate cancer cell
sarcoma180 mouse sarcoma cell
SEM-1 human testicular seminoma cell
SGC-7901 human gastric adenocarcinoma cell
SK-MEL-28 human melanoma cell
SKOV-3 human ovarian cancer cell
SKOV-3/DDP cisplatin-resistant human ovarian cancer cell
SMMC human hepatocellular carcinoma cell
T-24 human bladder cancer cell
TCam-2 human testicular seminoma cell
U2-OS human osteosarcoma cell
U2-OS/Pt cisplatin-resistant human osteosarcoma cell
U87M human glioblastoma cell
WM1366 human melanoma cell

References
1. Shimada, M.; Itamochi, H.; Kigawa, J. Nedaplatin: A cisplatin derivative in cancer chemotherapy. Cancer Manag. Res. 2013,
5, 67–76. [CrossRef]
2. McKeage, M.J. Lobaplatin: A new antitumour platinum drug. Expert. Opin. Investig. Drugs 2001, 10, 119–128. [CrossRef]
3. Lee, K.H.; Hyun, M.S.; Kim, H.K.; Jin, H.M.; Yang, J.; Song, H.S.; Do, Y.R.; Ryoo, H.M.; Chung, J.S.; Zang, D.Y.; et al. Randomized,
multicenter, phase III trial of heptaplatin 1-h infusion and 5-fluorouracil combination chemotherapy comparing with cisplatin and
5-fluorouracil combination chemotherapy in patients with advanced gastric cancer. Cancer Res. Treat. 2009, 41, 12–18. [CrossRef]
4. Kenny, R.G.; Chuah, S.W.; Crawford, A.; Marmion, C.J. Platinum(IV) prodrugs—A step closer to Ehrlich’s vision? Eur. J. Inorg.
Chem. 2017, 2017, 1596–1612. [CrossRef]
5. Johnstone, T.C.; Suntharalingam, K.; Lippard, S.J. The next generation of platinum drugs: Targeted Pt(II) agents, nanoparticle
delivery, and Pt(IV) prodrugs. Chem. Rev. 2016, 116, 3436–3486. [CrossRef]
6. Dilrub, S.; Kalayd, G.V. Platinum-based drugs: Past, present and future. Cancer Chemother. Pharm. 2016, 77, 1103–1124. [CrossRef]
7. Wang, X.Y.; Guo, Z.J. Towards the rational design of platinum(II) and gold(III) complexes as antitumour agents. Dalton Trans.
2008, 1521–1532. [CrossRef] [PubMed]
Pharmaceuticals 2021, 14, 133 19 of 23

8. Argyriou, A.A.; Polychronopoulos, P.; Iconomou, G.; Chroni, E.; Kalofonos, H.P. A review on oxaliplatin-induced peripheral
nerve damage. Cancer Treat. Rev. 2008, 34, 368–377. [CrossRef] [PubMed]
9. McWhinney, S.R.; Goldberg, R.M.; McLeod, H.L. Platinum neurotoxicity pharmacogenetics. Mol. Cancer Ther. 2009, 8, 10–16.
[CrossRef]
10. Min, Y.Z.; Mao, C.Q.; Chen, S.M.; Ma, G.L.; Wang, J.; Liu, Y.Z. Combating the drug resistance of cisplatin using a platinum
prodrug based delivery system. Angew. Chem. Int. Ed. 2012, 51, 6742–6747. [CrossRef]
11. Wang, D.; Lippard, S.J. Cellular processing of platinum anticancer drugs. Nat. Rev. Drug Discov. 2005, 4, 307–320. [CrossRef]
12. Brabec, V.; Hrabina, O.; Kasparkova, J. Cytotoxic platinum coordination compounds. DNA binding agents. Coord. Chem. Rev.
2017, 351, 2–31. [CrossRef]
13. Usanova, S.; Piée-Staffa, A.; Sied, U.; Thomale, J.; Schneider, A.; Kaina, B.; Köberle, B. Cisplatin sensitivity of testis tumour cells is
due to deficiency in interstrand-crosslink repair and low ERCC1-XPF expression. Mol. Cancer 2010, 9, 248. [CrossRef] [PubMed]
14. Sakai, W.; Swisher, E.M.; Karlan, B.Y.; Agarwal, M.K.; Higgins, J.; Friedman, C.; Villegas, E.; Jacquemont, C.; Farrugia, D.J.; Couch,
F.J.; et al. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature 2008, 451, 1116–1120.
[CrossRef] [PubMed]
15. Johnstone, T.C.; Park, G.Y.; Lippard, S.J. Understanding and improving platinum anticancer drugs—Phenanthriplatin. Anticancer
Res. 2014, 34, 471–476. [PubMed]
16. Guo, W.J.; Zhang, Y.M.; Zhang, L.; Huang, B.; Tao, F.F.; Chen, W.; Guo, Z.J.; Xu, Q.; Sun, Y. Novel monofunctional platinum(II)
complex Mono-Pt induces apoptosis-independent autophagic cell death in human ovarian carcinoma cells, distinct from cisplatin.
Autophagy 2013, 9, 996–1008. [CrossRef]
17. Riddell, I.A.; Johnstone, T.C.; Park, G.Y.; Lippard, S.J. Nucleotide binding preference of the monofunctional platinum anticancer-
agent phenanthriplatin. Chem. Eur. J. 2016, 22, 7574–7581. [CrossRef]
18. Malina, J.; Farrell, N.P.; Brabec, V. DNA condensing effects and sequence selectivity of dna binding of antitumor noncovalent
polynuclear platinum complexes. Inorg. Chem. 2014, 53, 1662–1671. [CrossRef]
19. Johnstone, T.C.; Lippard, S.J. The chiral potential of phenanthriplatin and its influence on guanine binding. J. Am. Chem. Soc.
2014, 136, 2126–2134. [CrossRef]
20. Johnstone, T.C.; Alexander, S.M.; Lin, W.; Lippard, S.J. Effects of monofunctional platinum agents on bacterial growth: A
retrospective study. J. Am. Chem. Soc. 2014, 136, 116–118. [CrossRef]
21. Lovejoy, K.S.; Serova, M.; Bieche, I.; Emami, S.; D’Incalci, M.; Broggini, M.; Erba, E.; Gespach, C.; Cvitkovic, E.; Faivre, S.; et al.
Spectrum of cellular responses to pyriplatin, a monofunctional cationic antineoplastic platinum(II) compound, in human cancer
cells. Mol. Cancer Ther. 2011, 10, 1709–1719. [CrossRef]
22. Zhou, W.; Almeqdadi, M.; Xifaras, M.E.; Riddell, I.A.; Yilmaz, Ö.H.; Lippard, S.J. The effect of geometric isomerism on the
anticancer activity of the monofunctional platinum complex trans-[Pt(NH3 )2 (phenanthridine)Cl]NO3 . Chem. Commun. 2018, 54,
2788–2791. [CrossRef]
23. Johnstone, T.C.; Wilson, J.J.; Lippard, S.J. Monofunctional and higher-valent platinum anticancer agents. Inorg. Chem. 2013, 52,
12234–12249. [CrossRef] [PubMed]
24. Macquet, J.P.; Butour, J.L. Platinum-amine compounds: Importance of the labile and inert ligands for their pharmacological
activities toward L1210 leukemia cells. J. Natl. Cancer Inst. 1983, 70, 899–905. [PubMed]
25. Cleare, M.J.; Hoeschele, J.D. Antitumor activity of group VIII transition metal complexes. I. Platinum(II) complexes. Bioinorg.
Chem. 1973, 2, 187–210. [CrossRef]
26. Sundquist, W.I.; Bancroft, D.P.; Lippard, S.J. Synthesis, characterization, and biological activity of cis-diammineplatinum(II)
complexes of the DNA intercalators 9-aminoacridine and chloroquine. J. Am. Chem. Soc. 1990, 112, 1590–1596. [CrossRef]
27. Hollis, L.S.; Sundquist, W.I.; Burstyn, J.N.; Heiger-Bernays, W.J.; Bellon, S.F.; Ahmed, K.J.; Amundsen, A.R.; Stern, E.W.; Lippard,
S.J. Mechanistic studies of a novel class of trisubstituted platinum(II) antitumor agents. Cancer Res. 1991, 51, 1866–1875.
28. Zhang, S.; Lovejoy, K.S.; Shima, J.E.; Lagpacan, L.L.; Shu, Y.; Lapuk, A.; Chen, Y.; Komori, T.; Gray, J.W.; Chen, X.; et al. Organic
cation transporters are determinants of oxaliplatin cytotoxicity. Cancer Res. 2006, 66, 8847–8857. [CrossRef]
29. Lovejoy, K.S.; Todd, R.C.; Zhang, S.; McCormick, M.S.; D’Aquino, J.A.; Reardon, J.T.; Sancar, A.; Giacomini, K.M.; Lippard, S.J.
cis-Diammine(pyridine)chloroplatinum(II), a monofunctional platinum(II) antitumor agent: Uptake, structure, function, and
prospects. Proc. Natl. Acad. Sci. USA 2008, 105, 8902–8907. [CrossRef]
30. Wang, D.; Zhu, G.Y.; Huang, X.H.; Lippard, S.J. X-ray structure and mechanism of RNA polymerase II stalled at an antineoplastic
monofunctional platinum-DNA adduct. Proc. Natl. Acad. Sci. USA 2010, 107, 9584–9589. [CrossRef]
31. Park, G.Y.; Wilson, J.J.; Song, Y.; Lippard, S.J. Phenanthriplatin, a monofunctional DNA-binding platinum anticancer drug
candidate with unusual potency and cellular activity profile. Proc. Natl. Acad. Sci. USA 2012, 109, 11987–11992. [CrossRef]
[PubMed]
32. Casini, A.; Reedijk, J. Interactions of anticancer Pt compounds with proteins: An overlooked topic in medicinal inorganic
chemistry. Chem. Sci. 2012, 3, 3135–3144. [CrossRef]
33. Kellinger, M.W.; Park, G.Y.; Chong, J.; Lippard, S.J.; Wang, D. Effect of a monofunctional phenanthriplatin-DNA adduct on RNA
polymerase II transcriptional fidelity and translesion synthesis. J. Am. Chem. Soc. 2013, 135, 13054–13061. [CrossRef] [PubMed]
34. Almaqwashi, A.A.; Zhou, W.; Naufer, M.N.; Riddell, I.A.; Yilmaz, Ö.H.; Lippard, S.J.; Williams, M.C. DNA intercalation facilitates
efficient DNA-targeted covalent binding of phenanthriplatin. J. Am. Chem. Soc. 2019, 141, 1537–1545. [CrossRef] [PubMed]
Pharmaceuticals 2021, 14, 133 20 of 23

35. Gregory, M.T.; Park, G.Y.; Johnstone, T.C.; Lee, Y.S.; Yang, W.; Lippard, S.J. Structural and mechanistic studies of polymerase η
bypass of phenanthriplatin DNA damage. Proc. Natl. Acad. Sci. USA 2014, 111, 9133–9138. [CrossRef] [PubMed]
36. Baruah, H.; Rector, C.L.; Monnier, S.M.; Bierbach, U. Mechanism of action of non-cisplatin type DNA-targeted platinum anticancer
agents: DNA interactions of novel acridinylthioureas and their platinum conjugates. Biochem. Pharm. 2002, 64, 191–200. [CrossRef]
37. Guddneppanavar, R.; Saluta, G.; Kucera, G.L.; Bierbach, U. Synthesis, biological activity, and DNA-damage profile of platinum
threading intercalator conjugates designed to target adenine. J. Med. Chem. 2006, 49, 3204–3214. [CrossRef] [PubMed]
38. Guddneppanavar, R.; Choudhury, J.R.; Kheradi, A.R.; Steen, B.D.; Saluta, G.; Kucera, G.L.; Day, C.S.; Bierbach, U. Effect of the
diamine nonleaving group in platinum-acridinylthiourea conjugates on DNA damage and cytotoxicity. J. Med. Chem. 2007, 50,
2259–2263. [CrossRef]
39. Smyre, C.L.; Saluta, G.; Kute, T.E.; Kucera, G.L.; Bierbach, U. Inhibition of DNA synthesis by a platinum−acridine hybrid agent
leads to potent cell kill in nonsmall cell lung cancer. ACS Med. Chem. Lett. 2011, 2, 870–874. [CrossRef]
40. Kalayda, G.V.; Jansen, B.A.J.; Wielaard, P.; Tanke, H.J.; Reedijk, J. Dinuclear platinum anticancer complexes with fluorescent
N,N’-bis(aminoalkyl)-1,4-diaminoanthraquinones: Cellular processing in two cisplatin-resistant cell lines reflects the differences
in their resistance profiles. J. Biol. Inorg. Chem. 2005, 10, 305–315. [CrossRef]
41. Kalayda, G.V.; Jansen, B.A.J.; Molenaar, C.; Wielaard, P.; Tanke, H.J.; Reedijk, J. Dinuclear platinum complexes with N,N’-
bis(aminoalkyl)-1,4-diaminoanthraquinones as linking ligands. Part II. Cellular processing in A2780 cisplatin-resistant human
ovarian carcinoma cells: New insights into the mechanism of resistance. J. Biol. Inorg. Chem. 2004, 9, 414–422. [CrossRef]
42. Jansen, B.A.J.; Wielaard, P.; Kalayda, G.V.; Ferrari, M.; Molenaar, C.; Tanke, H.J.; Brouwer, J.; Reedijk, J. Dinuclear platinum
complexes with N,N’-bis(aminoalkyl)-1,4-diaminoanthraquinones as linking ligands. Part I. Synthesis, cytotoxicity, and cellular
studies in gA2780 human ovarian carcinoma cells. J. Biol. Inorg. Chem. 2004, 9, 403–413. [CrossRef]
43. Wu, S.D.; Wang, X.Y.; Zhu, C.C.; Song, Y.J.; Wang, J.; Li, Y.Z.; Guo, Z.J. Monofunctional platinum complexes containing a
4-nitrobenzo-2-oxa-1,3-diazole fluorophore: Distribution in tumour cells. Dalton Trans. 2011, 40, 10376–10382. [CrossRef]
[PubMed]
44. Wu, S.D.; Zhu, C.C.; Zhang, C.L.; Yu, Z.; He, W.J.; He, Y.F.; Li, Y.Z.; Wang, J.; Guo, Z.J. In vitro and in vivo fluorescent imaging of
a monofunctional chelated platinum complex excitable using visible light. Inorg. Chem. 2011, 50, 11847–11849. [CrossRef]
45. Chen, Z.F.; Zhang, S.P.; Shen, L.; Zhu, Z.Z.; Zhang, J. Fluorescence imaging of a new monofunctional platinum(II) complex
containing a thioflavin-T (ThT)-based fluorophore. New J. Chem. 2015, 39, 1592–1596. [CrossRef]
46. Zhang, C.; Guan, R.L.; Liao, X.X.; Ouyang, C.; Rees, T.W.; Liu, J.P.; Chen, Y.; Ji, L.N.; Chao, H. A mitochondria-targeting dinuclear
Ir-Ru complex as a synergistic photoactivated chemotherapy and photodynamic therapy agent against cisplatin-resistant tumour
cells. Chem. Commun. 2019, 55, 12547–12550. [CrossRef]
47. Bonnet, S. Why develop photoactivated chemotherapy? Dalton Trans. 2018, 47, 10330–10343. [CrossRef] [PubMed]
48. Dabids, L.M.; Kleemann, B. Combating melanoma: The use of photodynamic therapy as a novel, adjuvant therapeutic tool.
Cancer Treat Rev. 2011, 37, 465–475.
49. Zhou, Z.J.; Song, J.B.; Nie, L.M.; Chen, X.Y. Reactive oxygen species generating systems meeting challenges of photodynamic
cancer therapy. Chem. Soc. Rev. 2016, 45, 6597–6626. [CrossRef]
50. Ethirajan, M.; Chen, Y.H.; Joshi, P.; Pandey, R.K. The role of porphyrin chemistry in tumor imaging and photodynamic therapy.
Chem. Soc. Rev. 2011, 40, 340–362. [CrossRef] [PubMed]
51. Naik, A.; Rubbiani, R.; Gasser, G.; Spingler, B. Visible-light-induced annihilation of tumor cells with platinum-porphyrin
conjugates. Angew. Chem. Int. Ed. 2014, 53, 6938–6941. [CrossRef] [PubMed]
52. Yu, G.C.; Yu, S.; Saha, M.L.; Zhou, J.; Cook, T.R.; Yung, B.C.; Chen, J.; Mao, Z.W.; Zhang, F.W.; Zhou, Z.J.; et al. A discrete
organoplatinum(II) metallacage as a multimodality theranostic platform for cancer photochemotherapy. Nat. Commun. 2018, 9,
4335. [CrossRef]
53. Santoro, A.M.; Lo Giudice, M.C.; D’Urso, A.; Lauceri, R.; Purrello, R.; Milardi, D. Cationic porphyrins are reversible proteasome
inhibitors. J. Am. Chem. Soc. 2012, 134, 10451–10457. [CrossRef]
54. Bacellar, I.O.; Tsubone, T.M.; Pavani, C.; Baptista, M.S. Photodynamic efficiency: From molecular photochemistry to cell death.
Int. J. Mol. Sci. 2015, 16, 20523–20559. [CrossRef] [PubMed]
55. Tasso, T.T.; Tsubone, T.M.; Baptista, M.S.; Mattiazzi, L.M.; Acunha, T.V.; Iglesias, B.A. Isomeric effect on the properties of
tetraplatinated porphyrins showing optimized phototoxicity for photodynamic therapy. Dalton Trans. 2017, 46, 11037–11045.
[CrossRef]
56. Couto, G.K.; Pacheco, B.S.; Borba, V.M.; Junior, J.C.R.; Oliveira, T.L.; Segatto, N.V.; Seixas, F.K.; Acunha, T.V.; Iglesias, B.A.;
Collares, T. Tetra-cationic platinum(II) porphyrins like a candidate photosensitizers to bind, selective and drug delivery for
metastatic melanoma. J. Photoch. Photobio. B. 2020, 202, 111725. [CrossRef]
57. Hu, X.J.; Ogawa, K.; Li, S.; Kiwada, T.; Odani, A. A platinum functional porphyrin conjugate: An excellent cancer killer for
photodynamic therapy. Bull. Chem. Soc. Jpn. 2019, 92, 790–796. [CrossRef]
58. Xu, X.L.; Lin, F.W.; Du, Y.; Zhang, X.; Wu, J.; Xu, Z.K. Graphene oxide nanofiltration membranes stabilized by cationic porphyrin
for high salt rejection. Acs Appl. Mater. Interfaces 2016, 8, 12588–12593. [CrossRef] [PubMed]
59. Hu, X.J.; Ogawa, K.; Kiwada, T.; Odani, A. Water-soluble metalloporphyrinates with excellent photo-induced anticancer activity
resulting from high tumor accumulation. J. Inorg. Biochem. 2017, 170, 1–7. [CrossRef]
Pharmaceuticals 2021, 14, 133 21 of 23

60. Li, X.; Zheng, B.D.; Peng, X.H.; Li, S.Z.; Ying, J.W.; Zhao, Y.; Huang, J.D.; Yoon, J. Phthalocyanines as medicinal photosensitizers:
Developments in the last five years. Coord. Chem. Rev. 2017, 379, 147–160. [CrossRef]
61. Wonga, R.C.H.; Lo, P.C.; Ng, D.K.P. Stimuli responsive phthalocyanine-based fluorescent probes and photosensitizers. Coord.
Chem. Rev. 2017, 379, 30–46. [CrossRef]
62. Mitra, K.; Samsó, M.; Lyonsb, C.E.; Hartman, M.C.T. Hyaluronic acid grafted nanoparticles of a platinum(II)–silicon(IV)
phthalocyanine conjugate for tumor and mitochondria-targeted photodynamic therapy in red light. J. Mater. Chem. B 2018, 6,
7373–7377. [CrossRef] [PubMed]
63. Mao, J.F.; Zhang, Y.M.; Zhu, J.H.; Zhang, C.L.; Guo, Z.J. Molecular combo of photodynamic therapeutic agent silicon(IV)
phthalocyanine and anticancer drug cisplatin. Chem. Commun. 2009, 908–910. [CrossRef]
64. Raza, M.K.; Gautam, S.; Garai, A.; Mitra, K.; Kondaiah, P.; Chakravarty, A.R. Monofunctional BODIPY-appended imidazoplatin
for cellular imaging and mitochondria-targeted photocytotoxicity. Inorg. Chem. 2017, 56, 11019–11029. [CrossRef] [PubMed]
65. Wang, B.; Yuan, H.X.; Liu, Z.; Nie, C.Y.; Liu, L.B.; Lv, F.T.; Wang, Y.L.; Wang, S. Cationic oligo(p-phenylene vinylene) materials for
combating drug resistance of cancer cells by light manipulation. Adv. Mater. 2014, 26, 5986–5990. [CrossRef] [PubMed]
66. Xue, X.L.; Zhu, C.C.; Chen, H.C.; Bai, Y.; Shi, X.C.; Jiao, Y.; Chen, Z.Y.; Miao, Y.P.; He, W.J.; Guo, Z.J. A new approach to sensitize
antitumor monofunctional platinum(II) complexes via short time photo-irradiation. Inorg. Chem. 2017, 56, 3754–3762. [CrossRef]
67. Settembre, C.; Fraldi, A.; Medina, D.L.; Ballabio, A. Signals from the lysosome: A control centre for cellular clearance and energy
metabolism. Nat. Rev. Mol. Cell Biol. 2013, 14, 283–296. [CrossRef]
68. Bonam, S.R.; Wang, F.J.; Muller, S. Lysosomes as a therapeutic target. Nat. Rev. Drug Discov. 2019, 18, 923–948. [CrossRef]
[PubMed]
69. Klein, A.V.; Hambley, T.W. Platinum drug distribution in cancer cells and tumors. Chem. Rev. 2009, 109, 4911–4920. [CrossRef]
70. Xue, X.L.; Qian, C.G.; Fang, H.B.; Liu, H.K.; Yuan, H.; Guo, Z.J.; Bai, Y.; He, W.J. Photoactivated lysosomal escape of a
monofunctional PtII complex Pt-BDPA for nucleus access. Angew. Chem. Int. Ed. 2019, 58, 12661–12666. [CrossRef] [PubMed]
71. Liu, W.T.; Zhong, Y.F.; Liu, L.Y.; Shen, C.T.; Zeng, W.J.; Wang, F.Y.; Yang, D.Z.; Mao, Z.W. Solution structures of multiple
G-quadruplex complexes induced by a platinum(II)-based tripod reveal dynamic binding. Nat. Commun. 2018, 9, 3496. [CrossRef]
[PubMed]
72. Zheng, X.H.; Mu, G.; Zhong, Y.F.; Zhang, T.P.; Cao, Q.; Ji, L.N.; Zhao, Y.; Mao, Z.W. Trigeminal star-like platinum complexes
induce cancer cell senescence through quadruplex-mediated telomere dysfunction. Chem. Commun. 2016, 52, 14101–14104.
[CrossRef] [PubMed]
73. Zhong, Y.F.; Zhang, H.; Liu, W.T.; Zheng, X.H.; Zhou, Y.W.; Cao, Q.; Shen, Y.; Zhao, Y.; Qin, P.Z.; Ji, L.N.; et al. A platinum(II)-based
photosensitive tripod as an effective photodynamic anticancer agent through DNA damage. Chem. Eur. J. 2017, 23, 16442–16446.
[CrossRef]
74. Cao, Q.; Li, Y.; Freisinger, E.; Qin, P.Z.; Sigel, R.K.O.; Mao, Z.W. G-quadruplex DNA targeted metal complexes acting as potential
anticancer drugs. Inorg. Chem. Front. 2017, 4, 10–32. [CrossRef]
75. Dumat, B.; Bordeau, G.; Faurel-Paul, E.; Mahuteau-Betzer, F.; Saettel, N.; Metge, G.; Fiorini-Debuisschert, C.; Charra, F.; Teulade-
Fichou, M.P. DNA switches on the two-photon efficiency of an ultrabright triphenylamine fluorescent probe specific of AT regions.
J. Am. Chem. Soc. 2013, 135, 12697–12706. [CrossRef] [PubMed]
76. Zhong, Y.F.; Zhang, H.; Mu, G.; Liu, W.T.; Cao, Q.; Tan, C.P.; Ji, L.N.; Mao, Z.W. Nucleus-localized platinum(II)–triphenylamine
complexes as potent photodynamic anticancer agents. Inorg. Chem. Front. 2019, 6, 2817–2823. [CrossRef]
77. Wang, X.Y.; Guo, Z.J. Targeting and delivery of platinum-based anticancer drugs. Chem. Soc. Rev. 2013, 42, 202–224. [CrossRef]
78. Basu, U.; Banik, B.; Wen, R.; Pathak, R.K.; Dhar, S. The Platin-X series: Activation, targeting, and delivery. Dalton Trans. 2016, 45,
12992–13004. [CrossRef] [PubMed]
79. Kansara, M.; Teng, M.T.; Smyth, M.J.; Thomas, D.M. Translational biology of osteosarcoma. Nat. Rev. Cancer 2014, 14, 722–735.
[CrossRef]
80. Alpaslan, E.; Yazici, H.; Golshan, N.H.; Ziemer, K.S.; Webster, T.J. pH-dependent activity of dextran-coated cerium oxide
nanoparticles on prohibiting osteosarcoma cell proliferation. ACS Biomater. Sci. Eng. 2015, 1, 1096–1103. [CrossRef]
81. Reed, D.E.; Shokat, K.M. Targeting osteosarcoma. Proc. Natl. Acad. Sci. USA 2014, 111, 18100–18101. [CrossRef] [PubMed]
82. Whelan, J.S.; Davis, L.E. Osteosarcoma, chondrosarcoma, and chordoma. J. Clin. Oncol. 2018, 36, 188–193. [CrossRef] [PubMed]
83. Makris, G.; Tseligka, E.D.; Pirmettis, I.; Papadopoulos, M.S.; Vizirianakis, I.S.; Papagiannopoulou, D. Development and pharma-
cological evaluation of new bone-targeted 99mTc-radiolabeled bisphosphonates. Mol. Pharm. 2016, 13, 2301–2317. [CrossRef]
84. Zhang, Z.Q.; Wang, X.Y.; Luo, C.; Zhu, C.C.; Wang, K.; Zhang, C.L.; Guo, Z.J. Dinuclear platinum(II) complexes with bone-
targeting groups as potential anti-osteosarcoma agents. Chem. Asian J. 2017, 12, 1659–1667. [CrossRef] [PubMed]
85. Zhang, Z.Q.; Zhu, Z.Z.; Luo, C.; Zhu, C.C.; Zhang, S.R.; Zhang, C.L.; Guo, Z.J.; Wang, X.Y. A potential bone-targeting hypotoxic
platinum(II) complex with an unusual cytostatic mechanism toward osteosarcoma cells. Inorg. Chem. 2018, 57, 3315–3322.
[CrossRef]
86. Kenny, R.G.; Marmion, C.J. Toward Multi-targeted platinum and ruthenium drugs-a new paradigm in cancer drug treatment
regimens? Chem. Rev. 2019, 119, 1058–1137. [CrossRef] [PubMed]
87. Hamidi, H.; Ivaska, J. Every step of the way: Integrins in cancer progression and metastasis. Nat. Rev. Cancer 2018, 18, 533–548.
[CrossRef]
Pharmaceuticals 2021, 14, 133 22 of 23

88. Chatzisideri, T.; Thysiadis, S.; Katsamakas, S.; Dalezis, P.; Sigala, I.; Lazarides, T.; Nikolakaki, E.; Trafalis, D.; Gederaas, O.A.;
Lindgren, M.; et al. Synthesis and biological evaluation of a platinum(II)-c(RGDyK) conjugate for integrin-targeted photodynamic
therapy. Eur. J. Med. Chem. 2017, 141, 221–231. [CrossRef]
89. Zamora, A.; Gandioso, A.; Massaguer, A.; Buenestado, S.; Calvis, C.; Hernández, J.L.; Mitjans, F.; Rodríguez, V.; Ruiz, J.;
Marchán, V. Toward angiogenesis inhibitors based on the conjugation of organometallic platinum(II) complexes to RGD peptides.
ChemMedChem 2018, 13, 1755–1762. [CrossRef]
90. Wu, T.; Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017, 387, 61–68. [CrossRef]
91. Franich, A.A.; Živković, M.D.; Ilić-Tomić, T.; Ðord̄ević, I.S.; Nikodinović-Runić, J.; Pavić, A.; Janjić, G.V.; Rajković, S. New minor
groove covering DNA binding mode of dinuclear Pt(II) complexes with various pyridine-linked bridging ligands and dual
anticancer-antiangiogenic activities. J. Biol. Inorg. Chem. 2020, 25, 395–409. [CrossRef]
92. Martinez-Outschoorn, U.E.; Peiris-Pages, M.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer metabolism: A therapeutic perspective.
Nat. Rev. Clin. Oncol. 2017, 14, 11–31. [CrossRef]
93. Yang, Y.H.; Karakhanova, S.; Hartwig, W.; D’haese, J.G.; Philippov, P.P.; Werner, J.; Bazhin, A.V. Mitochondria and mitochondrial
ros in cancer: Novel targets for anticancer therapy. J. Cell. Physiol. 2016, 231, 2570–2581. [CrossRef] [PubMed]
94. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell
proliferation. Science 2009, 324, 1029–1033. [CrossRef]
95. Wallace, D.C. Mitochondria and cancer. Nat. Rev. Cancer. 2012, 12, 685–698. [CrossRef]
96. Marrachea, S.; Dhar, S. The energy blocker inside the power house: Mitochondria targeted delivery of 3-bromopyruvate. Chem.
Sci. 2015, 6, 1832–1845. [CrossRef]
97. Taylor, R.W.; Turnbull, D.M. Mitochondrial DNA mutations in human disease. Nat. Rev. Genet. 2005, 6, 389–402. [CrossRef]
[PubMed]
98. Bouwman, P.; Jonkers, J. The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance.
Nat. Rev. Cancer 2012, 12, 587–598. [CrossRef] [PubMed]
99. Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer
2013, 13, 714–726. [CrossRef]
100. Fulda, S.; Galluzzi, L.; Kroemer, G. Targeting mitochondria for cancer therapy. Nat. Rev. Drug Discov. 2010, 9, 447–464. [CrossRef]
101. Lomeli, N.; Di, K.J.; Czerniawski, J.; Guzowski, J.F.; Bota, D.A. Cisplatin-induced mitochondrial dysfunction is associated with
impaired cognitive function in rats. Free Radic. Biol. Med. 2017, 102, 274–286. [CrossRef]
102. Guo, Y.; He, Y.F.; Wu, S.D.; Zhang, S.R.; Song, D.F.; Zhu, Z.Z.; Guo, Z.J.; Wang, X.Y. Enhancing cytotoxicity of a monofunctional
platinum complex via a dual-DNA-damage approach. Inorg. Chem. 2019, 58, 13150–13160. [CrossRef]
103. Zhang, C.; Guan, R.L.; Liao, X.X.; Ouyang, C.; Liu, J.P.; Ji, L.N.; Chao, H. Mitochondrial DNA targeting and impairment by a
dinuclear Ir–Pt complex that overcomes cisplatin resistance. Inorg. Chem. Front. 2020, 7, 1864–1871. [CrossRef]
104. Zhu, Z.Z.; Wang, Z.H.; Zhang, C.L.; Wang, Y.J.; Zhang, H.M.; Gan, Z.J.; Guo, Z.J.; Wang, X.Y. Mitochondrion-targeted platinum
complexes suppressing lung cancer through multiple pathways involving energy metabolism. Chem. Sci. 2019, 10, 3089–3095.
[CrossRef]
105. Wang, K.; Zhu, C.C.; He, Y.F.; Zhang, Z.Q.; Zhou, W.; Muhammad, N.; Guo, Y.; Wang, X.Y.; Guo, Z.J. Restraining cancer cells
by dual metabolic inhibition with a mitochondrion-targeted platinum(II) complex. Angew. Chem. Int. Ed. 2019, 58, 4638–4643.
[CrossRef] [PubMed]
106. Imming, P.; Sinning, C.; Meyer, A. Drugs, Their targets and the nature and number of drug targets. Nat. Rev. Drug Discov. 2006, 5,
821–834. [CrossRef] [PubMed]
107. Ramsay, R.; Tipton, K. Assessment of enzyme inhibition: A review with examples from the development of monoamine oxidase
and cholinesterase inhibitory drugs. Molecules 2017, 22, 1192. [CrossRef] [PubMed]
108. Tonks, N.K. Protein tyrosine phosphatases: From genes, to function, to disease. Nat. Rev. Mol. Cell Biol. 2006, 7, 833–846.
[CrossRef]
109. Lu, L.P.; Zhu, M.L. Protein tyrosine phosphatase inhibition by metals and metal complexes. Antioxid. Redox Sign. 2014, 20,
2210–2224. [CrossRef]
110. Yuan, C.X.; Wang, W.R.; Wang, J.W.; Li, X.H.; Wu, Y.B.; Li, S.D.; Lu, L.P.; Zhu, M.L.; Xing, S.; Fu, X.Q. Potent and selective PTP1B
inhibition by a platinum(II) complex: Possible implications for a new antitumor strategy. Chem. Commun. 2020, 56, 102–105.
[CrossRef]
111. Zhang, D.W.; Yip, Y.M.; Li, L.B. In silico construction of HK2-VDAC1 complex and investigating the HK2 binding-induced
molecular gating mechanism of VDAC1. Mitochondrion 2016, 30, 222–228. [CrossRef]
112. Li, S.N.; Li, J.J.; Dai, W.Q.; Zhang, Q.H.; Feng, J.; Wu, L.W.; Liu, T.; Yu, Q.; Xu, S.Z.; Wang, W.W.; et al. Genistein suppresses
aerobic glycolysis and induces hepatocellular carcinoma cell death. Br. J. Cancer. 2017, 117, 1518–1528. [CrossRef]
113. Muhammad, N.; Tan, C.P.; Muhammad, K.; Wang, J.; Sadia, N.; Pan, Z.Y.; Ji, L.N.; Mao, Z.W. Mitochondria-targeting monofunc-
tional platinum(II)–lonidamine conjugates for cancer cell de-energization. Inorg. Chem. Front. 2020, 7, 4010–4019. [CrossRef]
114. Waghorn, P.A.; Jackson, M.R.; Gouverneur, V.; Vallis, K.A. Targeting telomerase with radiolabeled inhibitors. Eur. J. Med. Chem.
2017, 125, 117–129. [CrossRef]
Pharmaceuticals 2021, 14, 133 23 of 23

115. Paul, A.; Maji, B.; Misra, S.K.; Jain, A.K.; Muniyappa, K.; Bhattacharya, S. Stabilization and structural alteration of the G-
quadruplex DNA made from the human telomeric repeat mediated by Troger’s base based novel benzimidazole derivatives. J.
Med. Chem. 2012, 55, 7460–7471. [CrossRef] [PubMed]
116. Huang, G.B.; Chen, S.; Qin, Q.P.; Luo, J.R.; Tan, M.X.; Wang, Z.F.; Zou, B.Q.; Liang, H. Preparation of platinum(II) complexes with
naphthalene imide derivatives and exploration of their in vitro cytotoxic activities. Inorg. Chem. Commun. 2019, 104, 124–128.
[CrossRef]
117. Huang, G.B.; Chen, S.; Qin, Q.P.; Luo, J.R.; Tan, M.X.; Wang, Z.F.; Zou, B.Q.; Liang, H. In vitro and in vivo activity of novel
platinum(II) complexes with naphthalene imide derivatives inhibiting human non-small cell lung cancer cells. New J. Chem. 2019,
43, 8146–8152. [CrossRef]
118. Qin, Q.P.; Zou, B.Q.; Wang, Z.F.; Huang, X.L.; Zhang, Y.; Tan, M.X.; Wang, S.L.; Liang, H. High in vitro and in vivo antitumor
activities of luminescent platinum(II) complexes with jatrorrhizine derivatives. Eur. J. Med. Chem. 2019, 183, 111727. [CrossRef]
119. Zou, H.H.; Wang, L.; Long, Z.X.; Qin, Q.P.; Song, Z.K.; Xie, T.; Zhang, S.H.; Liu, Y.C.; Lin, B.; Chen, Z.F. Preparation of 4-([2,20 :60 ,200 -
terpyridin]-40 -yl)-N,N-diethylaniline NiII and PtII complexes and exploration of their in vitro cytotoxic activities. Eur. J. Med.
Chem. 2016, 108, 1–12. [CrossRef]
120. Sabbatini, M.; Zanellato, I.; Ravera, M.; Gabano, E.; Perin, E.; Rangone, B.; Osella, D. Pt(IV) bifunctional prodrug containing
2-(2-propynyl)octanoato axial ligand: Induction of immunogenic cell death on colon cancer. J. Med. Chem. 2019, 62, 3395–3406.
[CrossRef]
121. Wong, D.Y.Q.; Ong, W.W.F.; Ang, W.H. Induction of immunogenic cell death by chemotherapeutic platinum complexes. Angew.
Chem. Int. Ed. 2015, 54, 6483–6487. [CrossRef]
122. Huang, K.B.; Wang, F.Y.; Feng, H.W.; Luo, H.J.; Long, Y.; Zou, T.T.; Chan, A.S.C.; Liu, R.; Zou, H.H.; Chen, Z.F.; et al. An
aminophosphonate ester ligand-containing platinum(II) complex induces potent immunogenic cell death in vitro and elicits
effective anti-tumor immune responses in vivo. Chem. Commun. 2019, 55, 13066–13069. [CrossRef]
123. Al-Khayal, K.; Vaali-Mohammed, M.A.; Elwatidy, M.; Bin Traiki, T.; Al-Obeed, O.; Azam, M.; Khan, Z.; Abdulla, M.; Ahmad, R. A
novel coordination complex of platinum (PT) induces cell death in colorectal cancer by altering redox balance and modulating
MAPK pathway. Bmc Cancer 2020, 20, 685. [CrossRef] [PubMed]
124. Choroba, K.; Machura, B.; Raposo, L.R.; Małecki, J.G.; Kula, S.; Pajak, ˛ M.; Erfurt, K.; Maroń, A.M.; Fernandes, A.R. Platinum(II)
complexes showing high cytotoxicity toward A2780 ovarian carcinoma cells. Dalton Trans. 2019, 48, 13081–13093. [CrossRef]
125. Wang, F.Y.; Tang, X.M.; Wang, X.; Huang, K.B.; Feng, H.W.; Chen, Z.F.; Liu, Y.N.; Liang, H. Mitochondria-targeted platinum(II)
complexes induce apoptosis-dependent autophagic cell death mediated by ER-stress in A549 cancer cells. Eur. J. Med. Chem. 2018,
155, 639–650. [CrossRef] [PubMed]
126. Ferraro, G.; Marzo, T.; Infrasca, T.; Cilibrizzi, A.; Vilar, R.; Messori, L.; Merlino, A. A case of extensive protein platination: The
reaction of lysozyme with a Pt(II)–terpyridine complex. Dalton Trans. 2018, 47, 8716–8723. [CrossRef]
127. Li, Z.; Zhou, J.; Gan, Y.; Yin, Y.H.; Zhang, W.C.; Yang, J.F.; Tang, Y.X.; Dai, Y.B. Synthesis of a novel platinum(II) complex with
6,7-dichloro-5,8-quinolinedione and the study of its antitumor mechanism in testicular seminoma. J. Inorg. Biochem. 2019, 197,
110701. [CrossRef] [PubMed]
128. Icsel, C.; Yilmaz, V.T.; Cevatemre, B.; Aygun, M.; Ulukaya, E. Structures and anticancer activity of chlorido platinum(II)
saccharinate complexes with mono- and dialkylphenylphosphines. J. Inorg. Biochem. 2019, 195, 39–50. [CrossRef]
129. Mitra, K.; Basu, U.; Khan, I.; Maity, B.; Kondaiah, P.; Chakravarty, A.R. Remarkable anticancer activity of ferrocenyl-terpyridine
platinum(II) complexes in visible light with low dark toxicity. Dalton Trans. 2014, 43, 751–763. [CrossRef]
130. Zhang, J.Y.; Wang, X.Y.; Tu, C.; Lin, J.; Ding, J.; Lin, L.P.; Wang, Z.M.; He, C.; Yan, C.H.; You, X.Z.; et al. Monofunctional platinum
complexes showing potent cytotoxicity against human liver carcinoma cell line BEL-7402. J. Med. Chem. 2003, 46, 3502–3507.
[CrossRef]

You might also like