You are on page 1of 12

Journal of Inorganic Biochemistry 156 (2016) 133144

Contents lists available at ScienceDirect

Journal of Inorganic Biochemistry

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

Group 11 complexes with amino acid derivatives: Synthesis and


antitumoral studies
Lourdes Ortego a, Margarida Meireles b, Cornelia Kasper c, Antonio Laguna a,
M. Dolores Villacampa a,, M. Concepcin Gimeno a,
a
Departamento de Qumica Inorgnica, Instituto de Sntesis Qumica y Catlisis Homognea (ISQCH), CSIC-Universidad de Zaragoza, E-50009 Zaragoza, Spain
b
Universidade de Lisboa, Faculdade de Cincias, Departamento de Qumica e Bioqumica e Centro de Qumica e Bioqumica, Campo Grande, 1749-016, Lisboa, Portugal
c
Department of Biotechnology, Institute for Applied Microbiology, University of Material Resource and Life Science, Muthgasse 18, 1180, Vienna, Austria

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

Article history: Gold(I), gold(III), silver(I) and copper(I) complexes with modied amino acid esters and phosphine ligands have
Received 22 September 2015 been prepared in order to test their cytotoxic activity. Two different phosphine fragments, PPh3 and PPh2py
Received in revised form 15 December 2015 (py = pyridine), have been used. The amino acid esters have been modied by introducing an aromatic amine
Accepted 28 December 2015
as pyridine that coordinates metal fragments through the nitrogen atom, giving complexes of the type
Available online 30 December 2015
[M(L)(PR3)]+ or [AuCl3(L)] (L = L-valine-N-(4-pyridylcarbonyl) methyl ester (L1), L-alanine-N-(4-pyridylcarbonyl)
Keywords:
methyl ester (L2), L-phenylalanine-N-(4-pyridylcarbonyl) methyl-ester) (L3); M = Au(I), Ag(I), Cu(I), PR3 = PPh3,
Antitumor properties PPh2py). The in vitro cytotoxic activity of metal complexes was tested against four tumor human cell lines and one
Gold(I) tumor mouse cell line. A metabolic activity test (3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide,
Silver(I) MTT) was used and IC50 values were compared with those obtained for cisplatin. Several complexes displayed
Copper(I) signicant cytotoxic activities. In order to determine whether antiproliferation and cell death are associated with
Amino acid derivatives apoptosis, NIH-3T3 cells were exposed to ve selected complexes (Annexin V+ FITC, PI) and analyzed by ow
Apoptosis cytometry. These experiments showed that the mechanism by which the complexes inhibit cell proliferation induc-
ing cell death in NIH-3T3 cells is mainly apoptotic.
2015 Elsevier Inc. All rights reserved.

1. Introduction with amino acids have been reported [22,23]. We have previously
reported on the synthesis and functionalization of gold(I) phosphine
In the last decades chemotherapeutic research based on metal nicotinic acid thiolate with amino acids providing complexes with
complexes has increased considerably, since the good results with very good cytotoxic activity [24,25].
platinum(II) compounds as cisplatin, the most widely used metal- Silver(I) complexes were used as therapeutic compounds from
based antitumor drug. Gold complexes have been largely studied and the middle ages mainly as antimicrobial agents. They have recently
have gained growing interest because of their high cell growth inhibi- regained attention as promising antitumor drugs, as shown in recent re-
tion [14]. Gold(I) complexes with several ligands such as phosphines, views [26,27]. Several silver(I) derivatives with O-, N-, P- or S- donor li-
thiolates, N-heterocyclic carbenes [59], or gold(III) species [1014] gands or N-heterocyclic carbene ligands have shown high cytotoxic
have been studied for their cytotoxic activities. Many of these deriva- behavior against diverse human tumor cells [2834]. Mechanisms of
tives have shown promising cytotoxic activities, in some cases overcom- action of silver(I) derivatives are not yet clear. However interactions of
ing cisplatin resistance to specic cancer cells. In addition they have the complexes with DNA and thiol groups of the proteins have been
shown a mechanism of action clearly different from that of platinum observed [26,27].
drugs. Several targets have been identied for gold complexes; the inhi- Copper is an essential trace metal for organisms. This plays an im-
bition of thioredoxine reductase being one of the more important to portant role to catalyze metabolic oxidations and transfer electrons in
date [2,1521]. In spite of the great number of reported gold complexes some enzymes as superoxide dismutase or cytochrome-c oxidase. The
which show activity in the biological systems, not many gold derivatives international agency for research on cancer has not classied copper
as a human carcinogen. Copper complexes have been investigated as
potential antitumor agents because they may be less toxic for normal
cells than for cancerous ones. However, as all essential trace metals,
Corresponding authors.
copper become toxic at slightly higher concentrations than the optimal
E-mail addresses: dvilla@unizar.es (M.D. Villacampa), gimeno@unizar.es level. Principal toxicity may be the result of its redox activity (Cu(I)/
(M.C. Gimeno). Cu(II)) and its afnity for heteroatoms of proteins or other

http://dx.doi.org/10.1016/j.jinorgbio.2015.12.018
0162-0134/ 2015 Elsevier Inc. All rights reserved.
134 L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144

biomolecules. An important number of copper complexes, specially 2. Results and discussion


copper(II), have been screened for their antitumor properties. They
have shown encouraging perspectives and their mechanism of action 2.1. Synthesis and spectroscopic characterization
has been investigated [35]. Although copper(I) complexes have been
less studied compared to copper(II), an important number of them Amino acid derivatives L1L3 were synthesized by reaction of
have showed potent cytotoxic activity. Copper(I) complexes with P- commercially isonicotinic acid chloride with the corresponding
donor ligands [3641] or N-heterocyclic carbene ligands [42] have amino acid methyl ester compounds, in the presence of
shown strong cytotoxic activities. Research has indicated possible dif- triethylamine to allow the condensation reaction (Scheme 1), by
ferences (in the mechanisms of action) between copper complexes following literature precedents [43,44]. Spectroscopic information
and antitumor platinum derivatives. is shown in the Experimental section.
Synthesis of gold, silver and copper complexes with modied amino The coordination chemistry of these ligands towards group 11
acid groups could be interesting in view of the possible biological prop- metal complexes has been studied. Gold(I) compounds 16 have
erties. The amino acids can serve as good carriers to deliver the metallic been prepared by reaction of L1L3 with an equimolecular amount
atoms to the biological target. In cancerous cells the amino acid recep- of the corresponding metal phosphine complexes [Au(OTf)(PPh3)]
tors are overexpressed and consequently the complexes could be or [Au(OTf)(PPh2py)] (OTf = CF3SO3, triuoromethanesulfonate),
more selective to abnormal cells. Therefore, it was thought that it whereas the gold(III) species 79 have been obtained by the reaction
would be a good idea to try to introduce metallic ions into the tumor of the ligands with AuCl3nH2O (Scheme 2).
cells by amino acid derivatives as ligands, to facilitate the cell perme- In the same manner, the reaction of L1L3 with the silver or copper
ation. In addition, it was considered that these ligands would be more complexes [Ag(OTf)(PR3)n] (n = 1, 2) or [Cu(NO3)(PPh3)2] has afforded
biocompatible and therefore reduce side effects in vivo. complexes 1018 and 1921, respectively (Scheme 3).
L1L3 ligands and the corresponding meta piridyl-aminoacid deriv- They have been characterized by means of IR, elemental analysis and
atives (Scheme 1) were previously prepared to synthesize a series of NMR spectroscopy. Assignments of the 1H NMR and 13C NMR signals
fac tricarbonyl rhenium diimine complexes [43]. The amino acid methyl were made on the basis of 2D 1H COSY and HSQC spectra. Complete
esters derivatives were modied by introducing a pyridine group which spectroscopic information of the complexes has been collected in the
enhances the ability to coordinate metal atoms. Their applications in Experimental section.
uorescent microscopy cell imaging in the MCF-7 cell line were report- The 1H NMR spectra of the compounds in acetone-d6 presented the
ed. These derivatives showed a good uptake but with a markedly expected resonances for all the protons (see Experimental section).
different effect on the cells. Interestingly, after irradiation, rhenium They showed, in Au(I), Au(III) and Ag(I), a low-eld shift of the pyridine
complexes with para amino acid derivatives induced damage in the protons H(2) and H(3), the nearest to the nitrogen atom, compared to
cell structure and cell death was observed. The cells treated with the those found in the free ligands, that conrms the coordination through
complexes with meta amino acid derivatives remained healthy. the pyridine nitrogen atom to the metal. The 13C{1H} NMR also showed
This caused us to consider the hypothesis that coinage metal com- a low-eld shift of the pyridine carbons C(2) and C(3). 1H NMR spectra
plexes synthesized with the para and meta substituted pyridine amino of Cu(I) complexes 19 (L1), 20 (L2) and 21 (L3) showed the resonances
acid derivatives could have interesting cytotoxic properties. Here, we for the corresponding amino ester derivative without no signicant
report on the synthesis, structural characterization and on the study of changes to those observed in the free amino acid ester ligands. The
the biological activity of several group 11 metal complexes with the phenyl resonances indicated that only one phosphine group is coordi-
para derivatives. Unfortunately, it was not possible to measure the nated to the copper metal in each compound.
cytotoxic activity of gold(I) derivatives with the meta pyridyl-amino The 31P{1H} NMR spectra of gold(I) and copper(I) compounds con-
acid ligands, due to their poor stability in solution at room temperature. sist on one singlet from the phosphorus atom coordinated to the
The complexes decomposed to metallic gold in a few hours. The in vitro metal. The silver complexes with PPh3 are uxional in solution. Their
31 1
cytotoxic activities of metal complexes were tested against four tumor P{ H} NMR spectra at room temperature display one broad resonance
human cell lines, A-549 (human lung), Hep-G2 (human liver cancer), for complexes 11, 14, 16 and 17 or two broad resonances for complexes
HeLa (human cervix epithelial carcinoma), MCF-7 (breast cancer) and 10 and 13. This kind of uxionality is typical in the coordination chem-
one tumor mouse cell line, NIH-3T3 (mouse broblast) showing strong istry of silver and can be attributed to exchange phenomena involving
antitumor activity in vitro. Further experiments have been carried out in the neutral ligands. In the low temperature spectra the expected com-
order to determine the mechanism for which the complexes inhibit cell plex in each reaction was identied as the major product. At 203 K the
31 1
proliferation and cause cell death. P{ H} NMR spectra of the mono-phosphine complexes 10, 13 and 16

Scheme 1. Depiction of selected ligands and their precursor.


L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144 135

Scheme 2. Synthesis of the gold(I) and gold(III) complexes with L1L3: i) [Au(OTf)(PR3)] and ii) AuCl3nH2O.

consist of two doublets due to coupling with 107Ag and 109Ag nuclei monoclinic space group P21/c with two and one molecules by asymmet-
(J107AgP around 600 Hz, J109AgP around 700 Hz) that correspond to ric unit, respectively (Figs. 3 and 4). The gold atoms in these complexes
the main derivative, [Ag(L)(PPh3)](OTf), and two doublets (J107AgP show a slightly distorted square-planar AuCl3N coordination. The angles
around 450 Hz, J109AgP around 550 Hz) assigned to the corresponding around gold show slight deviations from the ideal 90. The gold(III)
bis(phosphine) species [Ag(L)(PPh3)2](OTf) (Fig. 1). atoms are practically in the same plane generated by the three chloride
At 203 K the 31P{1 H} NMR spectra of the bis(phosphine) com- atoms and the pyridine nitrogen (average metal center deviation from
plexes 11, 14 and 17 show two doublets for the main derivative the planes formed by the four donor atoms is 0.010 ). As expected,
[Ag(L)(PPh3 )2](OTf), as the most intense resonances, and also as the asymmetric carbon atoms C7 in 7 and 8 retained the S conguration
minor components two doublets for the mono(phosphine), of the commercial amino acid methyl ester used in the synthesis of L1
[Ag(L)(PPh 3 )](OTf), and two doublets (J 107 AgP around 250 Hz, and L2.
J109AgP around 300 Hz) that can be assigned to the tris(phosphine),
[Ag(L)(PPh3 ) 3 ](OTf), derivatives. An equilibrium between phos- 2.3. Absorption and emission spectra
phine derivatives can be proposed as has been observed in other sil-
ver complexes [4550]. The 31P{1H} NMR spectra of complexes 12, UVvis spectra for the ligands and the complexes were recorded at
15 and 18 at r.t. show a broad resonance, that splits into a slightly ca. 105 M concentration in dichloromethane solution. The absorption
broad 6 symmetric lines at 203 K (Fig. 2). These spectra do not spectra of the ligands display one strong absorption at 230 nm and a
match with a phosphorus atom coupled to both silver isotopomers, weaker one at 263 nm. The spectra for all the complexes resemble
were only two doublets would appear. It resembles that of the those of the ligands with a slight variation in the energy, the strongest
starting material, which has a dinuclear structure, [Ag2 (OTf) 2(- absorption has a maximum at ca. 235 nm and a weaker absorption at
PPh2py)2], with bridging P,N units forming an eight membered ring ca. 265 nm. Those complexes bearing phosphine ligands, especially
[51], and corresponds to an AB system for the phosphorus atoms PPh2py display more intense and dened absorptions. As these absorp-
coupled to both silver nuclei with different coupling constants. For tions are present in the ligands and in all the complexes, including those
this reason we proposed a similar structure for the PPh 2 py com- with no phosphine ligands, we attributed them to intraligand * or
plexes, in which the modied pyridil amino ester ligand would coor- n * transition characteristic of aromatic systems.
dinate to each silver atom of the dinuclear unit (Fig. 2). The gold and silver complexes bearing 2-diphenylphosphino pyri-
dine ligands are emissive in solution and in the solid state, the date is
2.2. Crystal structure determination summarized in Table 1. In dichloromethane solutions the gold com-
plexes, 2, 4 and 6, and the silver derivatives, 12, 15 and 18, show an
The crystal structures of gold(III) complexes 7 and 8 have been de- emission around 370 and 400 nm, respectively. The same behavior is
termined by X-ray diffraction studies. They crystallized in the observed in the solid state, where the complexes display intense

Scheme 3. Synthesis of the silver(I) and copper(I) complexes with L1L3: i) [Ag(OTf)(PR3)n] and ii) [Cu(NO3)(PPh3)2].
136 L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144

Fig. 1. 31P{1H} NMR (CD3)2CO spectrum of [Ag(L1)(PPh3)](OTf) (10) at 203 K.

emissions at about 440 nm. In both, solution and solid state, these emis- silver(I) and copper(I) complexes appeared to be quite effective as cyto-
sions are assigned to intraligand transitions modied by coordination to toxic agents in vitro against the growth of the cancer cell lines tested.
the metal center. In these sense a redshift is observed on going from The gold(I) complexes showed in general to be effective as cytotoxic
gold to silver species in solutions, whereas in the solid state this redshift agents against the in vitro growth of all the tested cancer cell lines. They
is only noticeable in complex 18. are particularly effective against HeLa (IC50 values range between 4.33
and 11.71 M) and MCF-7 (IC50 values range between 0.45 and
20.05 M) cell lines being 2, 4 and 5 the most potent agents against
2.4. Biological evaluation MCF-7 with IC50 values of 0.65, 1.78 and 0.45 M, respectively. The var-
iation of the tertiary phosphine from PPh3 to PPh2py provided higher
2.4.1. Cytotoxic studies cytotoxicity. Also if we consider the amino acid ester derivative in the
The in vitro cytotoxic activity of metal complexes (except complexes pyridine moiety, the lower IC50 values have been achieved with the va-
11, 14, 17) was tested against four tumor human cell lines, A-549 line methyl ester.
(human lung), Hep-G2 (human liver cancer), HeLa (human cervix epi- Silver(I) derivatives presented globally lower activities in all the
thelial carcinoma), MCF-7 (breast cancer), and one tumor mouse cell cancer cell lines compared with those of gold(I). Again an increase in cy-
line, NIH-3T3 (mouse broblast), and IC50 values compared with those totoxicity is achieved when changing the phosphine from PPh3 to
of cisplatin [52,53]. As control, metal-free ligands L1, L2 and L3 were PPh2py. Specially, complex 18 with PPh2py is a powerful agent against
tested for cytotoxicity against all the cell lines except the MCF-7 cells. the HeLa cell line (IC50 value of 3.40 M), and the species 16,
Compounds are not soluble in water, but they are soluble in DMSO [Ag(L3)(PPh3)](OTf), and 18, [Ag(L3)(PPh2py)](OTf), showed strong
and in the DMSO/water mixtures used in the cytotoxicity tests, which cytotoxic activity with IC50 values of 5.63 and 2.81 M, respectively,
contain a small amount of DMSO (5%). We did not observe any precip- against MCF-7 cell line. Considering the amino acid ester derivative in
itation of the complexes while performing the tests. The colorless the pyridine moiety, the lower IC50 values have been achieved with
(Au(I), Ag(I) Cu(I)) or yellow Au(III)-DMSO solutions are very stable the phenyl alanine methyl ester.
at room temperature for weeks. Copper(I) complexes also showed remarkable activities against NIH-
Cells were exposed to each compound for 48 h. By using the colori- 3T3, HeLa and MCF-7 cell lines with IC50 values ranging between 5.27
metric mitochondrial function-based MTT viability assay the IC50 values and 24.83 M. In this case and considering the amino acid ester deriva-
(nal concentration 5% DMSO) were calculated from doseresponse tive in the pyridine moiety, the lower IC50 values have been achieved
curves obtained by nonlinear regression analysis. IC50 values are con- with the valine and phenyl alanine methyl esters.
centrations of a drug required to inhibit tumor cell proliferation by In summary, almost all the gold(I) complexes are cytotoxic against
50%, compared to the control viability. Controls were performed using all the cell lines, being the stronger agents the PPh2py derivatives. In
cells grown in medium with 5% DMSO. IC50 values at 48 h are listed in general, a lower activity was observed for the silver(I) complexes
Table 2. although some of them showed high antiproliferative activity, particu-
As can be seen in Table 2, the tested ligands and the gold(III) larly complexes with the L3 (phenyl alanine methyl esther) ligand.
complexes showed no cytotoxic activity. By contrast, some gold(I), Also, a signicant antiproliferative activity can be observed for

Fig. 2. A) Proposed structures for complexes 12, 15 and 18 based on the structure of the compound [Ag2(OTf)2(-PPh2py)2]; B) 31P{1H}NMR spectrum (300 MHz, (CD3)2CO, 203 K) of
complex 15.
L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144 137

Fig. 3. Diagram of complex 7 (the second molecule within the asymmetric unit is omitted for clarity). Hydrogen atoms (except NH and C7-H) are omitted for clarity. Selected bond lengths
[] and angles []: Au(1)Cl(1) 2.257 (8); Au(1) Cl(2) 2.273(8); Au(1) Cl(3) 2.281(8); Au(1)N(1) 2.04(3); Cl(1)Au(1)N(1) 90.2(9); Cl(1)Au(1)Cl(2) 90.7(3); Cl(2)Au(1)
Cl(3) 91.2(3); N(1)Au(1)Cl(3) 88.0(9).

copper(I) complexes especially against the NIH-3T3 cell line. Although indicative of late apoptosis or necrosis [5558]. To determine whether
it is not possible to establish a reliable relation between composition antiproliferation and cell death are associated with apoptosis, NIH-3T3
and activity, the measured data showed that the MCF-7 and HeLa cells cells were exposed for 24 h to IC50 concentrations of complexes 2
are very sensitive to the metallic complexes tested, with most of the (Au), 16 (Ag), 19 (Cu), 20 (Cu) or 21 (Cu). Annexin V labeled with the
IC50 values being lower than those found for cisplatin, the antitumoral uorophore FITC, and the uorochrome propidium iodide PI were
agent taken as reference. We hypothesize that these complexes may added and cells were analyzed by ow cytometry. Flow cytometry stud-
enter the cells, as could be observed for the rhenium complexes with ies allow us to know whether cells are dying via apoptosis or necrosis. In
these para-amino acid ester ligands that showed a signicant uptake Fig. 5, the lower-right panel corresponds to early apoptotic cells
in MCF-7 cells [43]. Moreover these para rhenium derivatives induced (Annexin V+ FITC), the upper-left panel to necrotic cells (PI) and the
damage in the cell structure and cell death was observed. In contrast, upper right panel to cells in a necrotic process but whose death started
cells incubated with the meta ligands remained healthy. Unfortunately, as apoptotic (late apoptosis, Annexin V + PI). NIH-3T3 cell studies
the biological activity of the meta gold(I) complexes could not be inves- showed that 80% of untreated cells were alive after incubation of 24 h
tigated due to their poor stability in solution at room temperature. in 5% DMSO (Fig. 5a). Incubation with the complexes at the concentra-
tions corresponding to the IC50 values showed that for complexes
2.4.2. Detection of cell death using FITC-conjugated annexin V (ow [Au(L1)(PPh2py)](OTf) (2) (Fig. 5b) and [Ag(L3)(PPh3)](OTf) (16)
cytometry) (Fig. 5c) 100% of cells were in 100% late apoptosis. Cells incubated
Apoptosis is the nal effect of the treatment with multiple with [Cu(L)(PPh3)](NO3) (L = L1, 19; L2, 20; L3, 21) (Fig. 5df) com-
chemotherapeutical agents in cancer. Many anticancer drugs (as cis- plexes were alive (2519%), in late apoptosis (5257%) and in early ap-
platin) are effective against cancerous cells by causing apoptosis by optosis (1623%). All these observations suggest that the mechanism by
the generation of reactive oxygen species (ROS) and DNA damage [15, which the complexes inhibit cell proliferation in NIH-3T3 cells inducing
28,54]. Apoptosis is a programmed mode of cell death that is accompa- cell death, is mainly apoptotic. Moreover, it can be observed that com-
nied by numerous morphological as well as biochemical changes in the plexes 1921 induced a slower cytotoxic mechanism, although they
cellular architecture that include cell surface changes. In apoptotic cells, are more effective cytotoxic agents (smaller IC50 values) than 2 and 16.
phosphatidylserine (PS) is translocated from the inner to the outer leaf-
let of the plasma membrane, where it can be detected by its binding to 3. Conclusions
the intracellular protein Annexin V, labeled with a uorophore. The
Annexin V assay is able to distinguish apoptotic cells from viable or ne- A series of gold, silver and copper complexes, [M(L)(PR3)]+ or
crotic cells on the basis of the resulting uorescence of the former cells. [AuCl3(L)] (L = L-valine-N-(4-pyridylcarbonyl) methyl ester (L1), L-ala-
The simultaneous use of a dead cell stain employs the uorochrome nine-N-(4-pyridylcarbonyl) methyl ester (L2), L-phenylalanine-N-
propidium iodide (PI), which can only penetrate dead or dying cells (4-pyridylcarbonyl) methyl ester (L3); M = Au(I), Ag(I), Cu(I),
that have lost or are losing membrane integrity. PI intercalates into PR3 = PPh3, PPh2py), with modied amino acid esters and phosphine
DNA to produce a highly uorescent adduct, whose observation is ligands have been prepared. The in vitro cytotoxic activity of metal

Fig. 4. Diagram of complex 8. Hydrogen atoms (except NH and C7-H) are omitted for clarity. Selected bond lengths [] and angles [] for complex 8: Au(1)Cl(1) 2.2838(11); Au(1) Cl
(2) 2.2599(10); Au(1) Cl(3) 2.2766(11); Au(1)N(1) 2.028(3); Cl(1)Au(1)N(1) 90.14(9); Cl(1)Au(1)Cl(2) 92.33(4); Cl(2)Au(1)Cl(3) 90.72(4); N(1)Au(1)Cl(3) 86.82(9).
138 L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144

Table 1 prepared according to published procedures. [Au(OTf)(PPh3)] or


Excitation and emission values for complexes 2, 4, 6, 12, 15 and 18. [Au(OTf)(PPh2py)] was obtained by reaction of [AuCl(PPh3)] [59] or
Solution (CH2Cl2 7 104 M) Solid [AuCl(PPh2py)] [60] with Ag(OTf) in dichloromethane and used in
Excitation (nm) Emission (nm) Excitation (nm) Emission (nm)
situ. All other reagents were commercially available and used without
further purication. C, H, and N analyses were carried out with a
2 330 370 395 440
PerkinElmer 2400 microanalyzer. 1H, 13C{1H} and 31P{1H}, including
4 320 376 390 440
6 320 365 395 440 2D experiments, were recorded at room temperature on a Bruker
12 337 390 392 440 Avance 400 and a Bruker ARX 300 spectrometers. Chemical shifts
15 337 390 390 440 (, ppm) were reported relative to the solvent peaks in the 1H, 13C
18 362 440 357 480 spectra or external 85% H3PO4 in 31P spectra. UVvis spectra were re-
corded with a 1 cm quartz cells on an Evolution 600 spectrophotometer.
Room temperature steady-state emission and excitation spectra were
complexes was tested against four tumor human cell lines and one recorded with a Jobin-Yvon-Horiba uorolog FL3-11 spectrometer
tumor mouse cell line. They were A-549 (human lung), Hep-G2 tted with a JY TBX picosecond detection module.
(human liver cancer), HeLa (human cervix epithelial carcinoma),
MCF-7 (breast cancer) and NIH-3T3 (mouse broblast). A metabolic ac-
tivity test (3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bro- 4.2. General synthesis of L1 L3
mide, MTT) was used and IC50 values were compared to cisplatin.
Almost all the gold(I) complexes are cytotoxic against all the cell lines, Isonicotinic acid chloride (4.08 g, 20 mmol), triethylamine (3.42 mL,
being the stronger agents the PPh2py derivatives. In general, a lower ac- 22 mmol) and the correspondent L-amino acid methyl ester (20 mmol)
tivity was observed for the silver(I) complexes although some of them were stirred in dry DCM for 24 h at room temperature under an argon
showed high antiproliferative activity, specially complexes with the L3 atmosphere. Then an aqueous solution of NaHCO3 was added to the
ligand. Also, a signicant cytotoxic activity can be observed for mixture and an extraction was performed (3 20 mL DCM). The com-
copper(I) complexes especially against the NIH-3T3 cell line. Although bined organic layers were dried over MgSO4 and ltered over celite.
it is not possible to establish a reliable relation between composition The volume was reduced to 10 mL, and addition of hexane and/or
and activity, the measured data showed that the MCF-7 and HeLa cells diethyl ether afforded L1 as orange solid, L2 as beige solid and L3 as a
are very sensitive to the metallic complexes tested, with most of the brown oil.
IC50 values being lower than those found for cisplatin, the reference
used. Flow cytometry studies of NIH-3T3 cells exposed to IC50 concen-
trations of ve selected complexes, showed that the mechanism by 4.3. L-Valine-N-(4-pyridylcarbonyl) methyl ester (L1)
which the complexes inhibit cell proliferation inducing cell death is
mainly apoptotic. Copper complexes tested induced a slower cytotoxic Yield: 4.30 g, 91%. Anal. Calcd. (%) for C12H16N2O3 (236.26): C, 61.00;
mechanism than gold and silver derivatives, in spite of being more H, 6.83; N, 11.86. Found: C, 61.12; H, 6.84; N, 11.95. IR (cm 1):
cytotoxic. (NH) = 3245, (COcarboxylate) = 1729, (COamide) = 1660. 1H NMR
(400 MHz, (CD3)2CO) : 8.71 (m, 2H, H2), 8.05 (s br, 1H, NH), 7.78 (m,
4. Experimental section 2H, H3), 4.57 (dd, 1H, NHCH, 3JHH 8.4 Hz, 3JHH 6.4 Hz), 3.72 (s, 3H,
COOCH3), 2.26 (m, 1H, CH(CH3)2), 1.02 (d, 3H, CH(CH3)2, 3JHH 6.8 Hz),
4.1. General procedures 1.00 (d, 3H, CH(CH3)2, 3JHH 6.8 Hz). 13C{1H}NMR (400 MHz, (CD3)2CO)
: 173.62 (s, 1C, COOCH3), 167.44 (s, 1C, CONH), 152.15 (s, 2C, C2),
The synthesis of the copper complexes 1921 were carried out 143.19 (s, 1C, C4), 123.17 (s, 2C, C3), 60.20 (s, 1C, NHCH), 53.19 (s, 1C,
under Ar atmosphere, in solvents dried and degassed prior to use. The COOCH3), 32.37 (s, 1C, CH(CH3)2), 20.45 (s, 1C, CH(CH3)2), 19.77 (s,
starting material [Ag(OTf)(PPh3)] [45] or [Ag(OTf)(PPh2py)] [51] was 1C, CH(CH3)2). UVvis, max nm: 230, 263.

Table 2
IC50 (48 h) values of complexes and ligands against tumor cells.

IC50 A-549 Hep-G2 NIH-3 T3 HeLa MCF-7

L Cisplatin 10.22 0.44 [52] 25 3.1 [53] nt 10 2.2 [52] 21.65 0.22 [51]
L1 N50 N50 N50 N50 nt
L2 N50 N50 N50 N50 nt
L3 N50 N50 N50 N50 nt
AuPPh3 1 24.56 0.34 nt 4.76 0.13 4.43 1.43 6.04 0.33
3 25.35 0.40 10.50 0.46 10.75 0.43 11.71 1.06 20.05 0.34
5 N50 25.95 0.16 3.71 0.64 10.66 0.33 0.45 0.20
AuPPh2py 2 10.21 0.60 10.18 0.41 13.41 1.37 4.33 0.46 0.65 0.34
4 25.22 0.59 10.81 0.78 10.31 0.37 5.58 0.86 1.78 0.05
6 25.35 0.44 10.85 0.54 6.43 1.55 6.93 0.50 10.14 0.69
AuCl3 7 N50 N50 N50 N50 nt
8 N50 N50 N50 N50 nt
9 N50 N50 N50 N50 nt
AgPPh3 10 25.22 0.37 10.50 0.45 24.92 0.07 48.91 0.13 nt
13 50 N50 10.08 0.14 25.78 0.82 nt
16 N50 25.18 0.36 10.59 0.22 13.98 1.69 5.63 0.93
AgPPh2py 12 25.16 0.34 5.47 0.27 25.07 0.25 N50 nt
15 N50 N50 25.31 0.18 N50 nt
18 N50 25.42 0.42 4.78 0.13 3.40 0.09 2.81 0.66
CuPPh3 19 24.50 0.35 35.12 0.34 1.82 0.30 8.13 1.05 12.94 0.75
20 24.56 0.32 25.02 0.38 1.98 0.32 24.83 2.70 5.63 0.93
21 10.47 0.23 25.17 0.23 2.12 0.38 5.27 1.07 10.55 0.45

nt: not tested.


L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144 139

Fig. 5. NIH-3T3 control cells grown for 24 h in 5% DMSO (a); NIH-3 T3 cells treated for 24 h with compounds 2 (b); 16 (c); 19 (d); 20 (e); or 21 (f) (concentrations corresponded to IC50
values). Apoptosis/necrosis hallmarks were analyzed as described in Experimental section. The percentage of cells in each region is indicated.

4.4. L-Alanine-N-(4-pyridylcarbonyl) methyl ester (L2) alanine-N-(4-pyridylcarbonyl) methyl ester (L1; 47 mg, 0.2 mmol) or
L-valine-N-(4-pyridylcarbonyl) methyl ester (L2; 42 mg, 0.2 mmol) or
Yield: 3.70 g, 89%. Anal. Calcd. (%) for C10H12N2O3 (208.21): C, 57.68, L-phenylalanine-N-(4-pyridylcarbonyl) methyl ester (L3; 64 mg,
H, 5.81, N, 13.45. Found: C, 57.42; H, 5.92; N, 13.46. IR (cm1): (NH) = 0.2 mmol) were stirred in DCM (20 mL) for 2 h at room temperature.
3296, (COcarboxylate) = 1731, (COamide) = 1645, sym(C = N) = 1537. The volume was reduced to 5 mL, and addition of hexane afforded com-
1
H NMR (400 MHz, (CD3)2CO) : 8.72 (m, 2H, H2), 8.22 (s br, 1H, NH), pounds 16. 1 and 3 as white solids, 2 and 4 as beige solids and 5 and 6
7.78 (m, 2H, H3), 4.66 (qd, 1H, NHCH, 3JHH 7.3 Hz, 3JHH 7.3 Hz), 3.70 (s, as brown oils.
3H, COOCH3), 1.48 (d, 3H, CHCH3, 3JHH 7.3 Hz). 13C{1H}NMR (400 MHz,
(CD3)2CO) : 174.39 (s, 1C, COOCH3), 166.61 (s, 1C, CONH), 152.08 (s,
2C, C2), 142.83 (s, 1C, C4), 122.87 (s, 2C, C3), 53.25 (s, 1C, NHCH), 50.31 4.6.1. [Au(L1)(PPh3)](OTf) (1)
(s, 1C, COOCH3), 18.27 (s, 1C, CHCH3). UVvis, max nm: 230, 263. Yield: 132 mg, 78%. Anal. Calcd. (%) for C31H31AuF3N2O6PS (844.59):
C, 44.09; H, 3.70; N, 3.32; S, 3.80. Found: C, 44.18; H, 3.55; N, 3.29; S,
3.63. IR (cm1): (NH) = 3290, (COcarboxylate) = 1737, (COamide) =
4.5. L-Phenylalanine-N-(4-pyridylcarbonyl) methyl ester (L3) 1666, sym(C_N) = 1537, asym(SO3) = 1252, sym(CF3) = 1221,
asym(CF3) = 1151, sym(SO3) = 1028. 1H NMR (300 MHz, (CD3)2CO)
Yield: 5.42 g, 95%. Anal. Calcd. (%) for C16H16N2O3 (284.31): C, 67.59; : 9.15 (m, 2H, H2), 8.40 (d, 1H, NH, 3JHH 8.2 Hz), 8.33 (m, 2H, H3),
H, 5.67; N, 9.85. Found: C, 67.17; H, 5.50; N, 9.81. IR (cm1): (NH) = 7.68 (m, 15H, HPh), 4.57 (m, 1H, NHCH), 3.72 (s, 3H, COOCH3), 2.29
3282, (COcarboxylate) = 1740, (COamide) = 1647, sym (C_N) = (m, 1H, CH(CH3)2), 1.03 (m, 6H, CH(CH3)2). 19F NMR (400 MHz,
1532. 1H NMR (400 MHz, (CD3)2CO) : 8.67 (m, 2H, H2), 8.39 (d, 1H, (CD3)2CO) : -78.82 (s, 1F, OTf). 31P{1H}NMR (400 MHz, (CD3)2CO) :
NH, 3JHH 7.9 Hz), 7.69 (m, 2H, H3), 7.27 (m, 5H, CH2Ph), 4.96 (m, 1H, 29.8 (s, 1P, AuPPh3). 13C{1H}NMR (300 MHz, (CD3)2CO) : 173.22 (s,
NHCH), 3.69 (s, 3H, COOCH3), A = 3.31 B = 3.17 (2H, system ABX, 1C, COOCH3), 165.59 (s, 1C, CONH), 154.14 (s, 2C, C2), 147.60 (s, 1C,
CH2Ph, JAX 5.4 Hz, JBX 9.4 Hz, JAB 13.9 Hz). 13C{1H}NMR (400 MHz, C4), 136.22 (d, 6C, CorthoPh, 2JCP 13.6 Hz), 136.56 (d, 3C, CparaPh, 4JCP
(CD3)2CO) : 173.46 (s, 1C, COOCH3), 167.03 (s br, 1C, CONH), 152.05 2.6 Hz), 131.58 (d, 6C, CmetaPh, 3JCP 12.1 Hz), 129.51 (d, 3C, CipsoPh, 1JCP
(s, 2C, C2), 142.88 (s, 1C, C4), 139.02 (s, 1C, CipsoCH2Ph), 130.95 (s, 2C, 66.4 Hz), 126.48 (s, 2C, C3), 60.72 (s, 1C, NHCH), 53.29 (s, 1C,
CorthoCH2Ph), 130.16 (s, 1C, CparaCH2Ph), 128.49 (s, 2C, CmetaCH2Ph), 122.93 COOCH3), 32.33 (s, 1C, CH(CH3)2), 20.43 (s, 1C, CH(CH3)2), 19.88 (s,
(s, 2C, C3), 56.17 (s, 1C, NHCH), 53.47 (s, 1C, COOCH3), 38.82 (s, 1C, 1C, CH(CH3)2). UVvis, max nm: 233, 265 (br).
CH2Ph). UVvis, max nm: 230, 263.
4.6.2. [Au(L1)(PPh2py)](OTf) (2)
4.6. Synthesis of [Au(L)(PR3)](OTf) (L1, R3_Ph3 (1), Ph2py (2); L2, R3_Ph3 Yield: 137 mg, 81%. Anal. Calcd. (%) for C30H30AuF3N3O6PS (845.58):
(3), Ph2py (4); L3, R3_Ph3 (5), Ph2py (6)) C, 42.61; H, 3.58; N, 4.97; S, 3.79. Found: C, 42.83; H, 3.42; N, 5.15; S,
3.93. IR (cm1): (NH) = 3287, (COcarboxylate) = 1731, (COamide) =
A solution of [Au(OTf)(PPh3)] (127.68 mg, 0.2 mmol) or 1661, sym(C_N) = 1536, asym(SO3) = 1253, sym(CF3) = 1221,
[Au(OTf)(PPh2py)] (128.67 mg, 0.21 mmol) prepared in situ and L- asym(CF3) = 1153, sym(SO3) = 1028. 1H NMR (400 MHz, (CD3)2CO)
140 L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144

: 9.04 (m, 1H, H6), 8.84 (m, 2H, H2), 8.20 (m, 1H, NH), 8.11 (m, 1H, H4), JAX 5.4Hz, JBX 9.4 Hz, JAB 13.9 Hz). 31P{1H}NMR (400 MHz, (CD3)2CO)
7.93 (m, 2H, H3), 7.66 (m, 12H, HPh, 3, 5), 4.56 (m, 1H, NHCH), 3.72 (s, : 29.5 (s br, 1P, AuPPh3). 13C{1H}NMR (300 MHz, (CD3)2CO) : 173.09
3H, COOCH3), 2.27 (m, 1H, CH(CH3)2), 1.02 (m, 6H, CH(CH3)2). 19F (s, 1C, COOCH3), 165.11 (s br, 1C, CONH), 154.06 (s, 2C, C2), 146.97 (s,
NMR (400 MHz, (CD3)2CO) : 80.06 (s, 1F, OTf). 31P{1H}NMR 1C, C4), 139.13 (s, 1C, CipsoCH2Ph), 136.17 (d, 6C, CorthoPh, 2JCP 13.6 Hz),
(400 MHz, (CD3)2CO) : 31.3 (s, 1P, AuPPh2py). 13C{1H}NMR 134.50 (d, 3C, CparaPPh3 4JCP 2.8 Hz), 131.54 (d, 6C, CmetaPh, 3JCP 12.1 Hz),
(400 MHz, (CD3)2CO) : 172.54 (s, 1C, COOCH3), 165.74 (s, 1C, CONH), 131.08 (s, 2C, CorthoCH2Ph), 130.24 (s, 1C, CparaCH2Ph), 128.58 (s, 2C,
153.59 (d, 1C, C6, 3JCP 16.8 Hz), 151.80 (s, 2C, C2), 143.84 (s, 1C, C4), CmetaCH2Ph), 129.58 (d, 3C, CipsoPh, 1JCP 61.0 Hz), 125.97 (s, 2C, C3), 56.60
139.56 (d, 1C, C4, 3JCP 9.0 Hz), 135.53 (d, 4C, CorthoPh, 2JCP 13.6 Hz), (s, 1C, NHCH), 53.54 (s, 1C, COOCH3), 38.76 (s, 1C, CH2Ph). UVvis,
133.68 (d, 2C, CparaPh, 4JCP 2.8 Hz), 132.16 (d, 1C, C3, 2JCP 25.2 Hz), max nm: 235, 265 (br).
130.53 (d, 4C, CmetaPPh, 3JCP 12.0 Hz), 127.63 (s, 1C, C5), 123.44 (s, 2C,
C3), 59.50 (s, 1C, NHCH), 52.30 (s, 1C, COOCH3), 31.36 (s, 1C,
CH(CH3)2), 19.51 (s, 1C, CH(CH3)2), 18.91 (s, 1C, CH(CH3)2). UVvis, 4.6.6. [Au(L3)(PPh2py)](OTf) (6)
max nm: 232, 265. Yield: 127 mg, 71%. Anal. Calcd. (%) for C34H30AuF3N3O6PS (893.62):
C, 45.70; H, 3.38; N, 4.70; S, 3.59. 1028. Found: C, 45.72; H, 3.44; N, 4.34;
S, 3.77. IR (cm1): (NH) = 3313, (COcarboxylate) = 1741, (COamide) =
1668, sym(C_N) = 1541, asym(SO3) = 1270 and 1251, sym(CF3) =
1222, asym(CF3) = 1151, sym(SO3) = 1028. 1H NMR (400 MHz,
(CD3)2CO) : 9.08 (s br, 1H, H6), 8.83 (s br, 2H, H2), 8.40 (s br, 1H,
NH), 8.13 (m, 1H, H4), 7.86 (m, 2H, H3), 7.65 (m, 12H, HPh, 3, 5), 7.28
(m, 5H, CH2Ph), 4.92 (m, 1H, NHCH), 3.71 (s, 3H, COOCH3), A = 3.30
4.6.3. [Au(L2)(PPh3)](OTf) (3) B = 3.16 (2H, system ABX, CH2Ph, JAX 5.4 Hz, JBX 9.4 Hz, JAB 13.9 Hz).
31 1
Yield: 160 mg, 98%. Anal. Calcd. (%) for C29H27AuF3N2O6PS (816.53): P{ H}NMR 400 MHz, (CD3)2CO) : 30.1 (s, 1P, AuPPh2py).
13 1
C, 42.66; H, 3.33; N, 3.43; S, 3.93. Found: C, 42.94; H, 3.54; N, 3.29; S, C{ H}NMR (400 MHz, (CD3)2CO) : 173.29 (s, 1C, COOCH3), 165.94
3.72. IR (cm1): (NH) = 3314, (COcarboxylate) = 1740, (COamide) = (s, 1C, CONH), 154.77 (d, 1C, H6, 1JCP 53.7 Hz), 152.77 (s, 2C, C2),
1666, sym(C_N) = 1539, asym(SO3) = 1268 and 1250, sym(CF3) = 144.87 (s br, 1C, C4), 140.77 (s, 1C, C4), 139.18 (s, 1C, CipsoCH2Ph),
1221, asym(CF3) = 1154, sym(SO3) = 1028. 1H NMR (400 MHz, 136.49 (d, 4C, CorthoPh, 2JCP 13.7 Hz), 134.72 (s br, 2C, CparaPh), 133.14
(CD3)2CO) : 9.10 (m, 2H, H2), 8.61 (s br, 1H, NH), 8.27 (m, 2H, H3), (d, 1C, C3, 2JCP 24.2 Hz), 131.53 (d, 4C, CmetaPh, 3JCP 12.2 Hz), 131.04 (s,
7.67 (m, 15H, HPh), 4.66 (qd, 1H, NHCH, 3JHH 7.3 Hz, 3JHH 7.3 Hz), 3.71 2C, CorthoCH2Ph), 130.23 (s, 1C, CparaCH2Ph), 128.73 (s br, 1C, C5), 128.56
(s, 3H, COOCH3), 1.51 (d, 3 H, CHCH3, 3JHH 7.3 Hz). 19F NMR (400 MHz, (s, 2C, CmetaCH2Ph), 124.39 (s, 2C, C3), 56.38 (s, 1C, NHCH), 53.51 (s, 1C,
(CD3)2CO) : 80.09 (s, 1F, OTf). 31P{1H}NMR (400 MHz, (CD3)2CO) COOCH3), 38.73 (s, 1C, CH2Ph). UVvis, max nm: 230, 265.
: 29.7 (s br, 1P, AuPPh3). 13C{1H}NMR (300 MHz, (CD3)2CO) : 174.18
(s, 1C, COOCH3), 165.63 (s, 1C, CONH), 153.82 (s, 2C, C2), 146.71 (s, 1C,
C4), 136.15 (d, 6C, CorthoPh, 2JCP 13.8 Hz), 134.45 (s, 3C, CparaPh), 131.53 4.7. Synthesis of [AuCl3(L)] (L1 (7), L2 (8), L3 (9))
(d, 6C, CmetaPh, 3JCP 12.1 Hz), 125.84 (s, 2C, C3), 53.46 (s, 1C, NHCH),
50.81 (s, 1C, COOCH3), 18.21 (s, 1C, CHCH3). UVvis, max nm: 235, To a methanol solution (25 mL) of L1 (47 mg, 0.2 mmol), L2 (42 mg,
265 (br). 0.2 mmol), or L3 (57 mg, 0.2 mmol) was added AuCl3 (47 mg,
0.2 mmol), at room temperature. After the addition, the reaction mix-
ture was stirred for 2 h. The volume was concentrated under vacuum
4.6.4. [Au(L2)(PPh2py)](OTf) (4)
to 5 mL and addition of distilled water gave yellow precipitates. The
Yield: 157 mg, 96%. Anal. Calcd. (%) for C28H26AuF3N3O6PS (817.53):
resulting yellow precipitates were ltered off and dried under vacuum.
C, 41.14; H, 3.21; N, 5.14; S, 3.92. Found: C, 41.55; H, 3.17; N, 4.84; S,
3.63. IR (cm1): (NH) = 3313, (COcarboxylate) = 1740, (COamide) =
1664, sym(C_N) = 1540, asym(SO3) = 1268 and 1250, sym 4.7.1. [AuCl3(L1)] (7)
(CF3) = 1221, asym(CF3) = 1155, sym(SO3) = 1028. 1H NMR Yield: 74 mg, 68%. Anal. Calcd. (%) for C12H16AuCl3N2O3 (539.59): C,
(400 MHz, (CD3)2CO) : 9.13 (s br, 1H, H6), 8.87 (m, 2H, H2), 8.42 (m, 26.71; H, 2.99; N, 5.19. Found: C, 27.17; H, 3.11; N, 5.27. IR (cm 1):
1H, NH), 8.15 (m, 1H, H4), 7.97 (m, 2H, H3), 7.76 (m, 12H, HPh, 3, 5), (NH) = 3276, (COcarboxylate) = 1743, (COamide) = 1645,
4.66 (qd, 1H, NHCH, 3JHH 7.3 Hz, 3JHH 7.3 Hz), 3.71 (s, 3H, COOCH3), (AuCl) = 363. 1H NMR (400 MHz, (CD3)2CO) : 9.30 (m, 2H, H2),
1.49 (d, 3H, CHCH3, 3JHH 7.3 Hz). 19F NMR (400 MHz, (CD3)2CO) : 8.43 (m, 1H, NH), 8.32 (m, 2H, H3), 4.62 (dd, 1H, NHCH, 3JHH 8.4 Hz,
80.07 (s, 1F, OTf). 31P{1H} NMR (400 MHz, (CD3)2CO) : 30.0 (s br, 3
JHH 6.4 Hz), 3.76 (s, 3H, COOCH3), 2.31 (m, 1H, CH(CH3)2), 1.05 (m,
AuPPh2py). 13C{1H}NMR (400 MHz, (CD3)2CO) : 174.34 (s, 1C, 6H, CH(CH3)2). 13C{1H}NMR (400 MHz, (CD3)2CO) : 173.08 (s, 1C,
COOCH3), 165.67 (s, 1C, CONH), 154.92 (d, 1C, C6, 3JCP 16.9 Hz), 154.08 COOCH), 165.34 (s, 1C, CONH), 153.01 (s, 2C, C2), 149.05 (s, 1C, C4),
(s, 1C, C2), 153.02 (s, 2C, C2), 144.97 (s, 1C, C4), 140.90 (d, 1C, C4, 3JHH 128.03 (s, 2C, C3), 60.54 (s, 1C, NHCH), 53.36 (s, 1C, COOCH3), 32.50 (s,
8.8 Hz), 136.51 (d, 4C, CorthoPh, 3JCP 13.7 Hz), 134.81 (s, 2C, CparaPh), 1C, CH(CH3)2), 20.40 (s, 1C, CH(CH3)2), 19.60 (s, 1C, CH(CH3)2). UV
133.16 (d, 1C, C3, 2JCP 23.7 Hz), 131.57 (d, 4C, CmetaPh, 4JCP 12.2 Hz), vis, max nm: 232, 265 (br).
128.83 (s, 1C, C5), 128.23 (s, 2C, CipsoPh, 3JCP 66.2 Hz), 124.59 (s, 2C,
C3), 53.44 (s, 1C, NHCH), 50.67 (s, 1C, COOCH3, 1C), 18.26 (s, 1C,
CHCH3). UVvis, max nm: 232, 265. 4.7.2. [AuCl3(L2)] (8)
Yield: 96 mg, 94%. Anal. Calcd. (%) for C10H12AuCl3N2O3 (511.54): C,
4.6.5. [Au(L3)(PPh3)](OTf) (5) 23.48; H, 2.36; N, 5.48. Found: C, 23.75; H, 2.38; N, 5.53. IR (cm 1):
Yield: 130 mg, 73%. Anal. Calcd. (%) for C35H31AuF3N2O6PS (892.63): (NH) = 3342, (COcarboxylate) = 1729, (COamide) = 1654,
C, 47.09; H, 3.50; N, 3.14; S, 3.59. Found: C, 46.99; H, 3.42; N, 2.93; S, (AuCl) = 363. 1H NMR (400 MHz, (CD3)2CO) : 9.29 (m, 2H, H2),
3.23. IR (cm1): (NH) = 3313, (COcarboxylate) = 1738, (COamide) = 8.63 (m, 1H, NH), 8.31 (m, 2H, H3), 4.69 (qd, 1H, NHCH, 3JHH 7.3 Hz,
3
1667, sym(C_N) = 1539, asym(SO3) = 1268 and 1251, sym(CF3) = JHH 7.3 Hz), 3.72 (s, 3H, COOCH3), 1.51 (d, 3H, CHCH3, 3JHH 7.3 Hz).
1222, asym(CF3) = 1151, sym(SO3) = 1028. 1H NMR (300 MHz, 13 1
C{ H}NMR (300 MHz, (CD3)2CO) : 174.02 (s, 1C, COOCH3), 164.48
(CD3)2CO) : 8.97 (m, 2H, H2), 8.51 (d, 1H, NH, 3JHH 8.0 Hz), 8.04 (m, (s, 1C, CONH), 153.16 (s, 2C, C2), 148.66 (s, 1C, C4), 127.80 (s, 2C, C3),
2H, H3), 7.66 (m, 15H, HPh), 7.28 (m, 5H, CH2Ph), 4.93 (m, 1H, NHCH), 53.58 (s, 1C, NHCH), 50.85 (s, 1C, COOCH3), 18.36 (s, 1C, CHCH3). UV
3.70 (s, 3H, COOCH3), A = 3.30 B = 3.16 (2H, system ABX, CH2Ph, vis, max nm: 232, 265 (br).
L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144 141

4.7.3. [AuCl3(L3)] (9) 4.8.3. [Ag(L2)(PPh3)](OTf) (13)


Yield: 97 mg, 83%. Anal. Calcd. (%) for C16H16AuN2O3Cl3 (587.63): C, Yield: 125 mg, 86%. Anal. Calcd. (%) for C29H27AgF3N2O6PS (727.44):
32.70; H, 2.74; N, 4.77. %. Found: C, 33.13; H, 2.50; N, 4.75. IR (cm1): C, 47.88; H, 3.74; N, 3.85; S, 4.41. Found: C, 47.52; H, 3.75; N, 3.90; S,
(NH) = 3342, (COcarboxylate) = 1737, (COamide) = 1653, 4.72. IR (cm1): (NH) = 3312, (COcarboxylate) = 1740, (COamide) =
(AuCl) = 363. 1H NMR (400 MHz, (CD3)2CO) : 9.26 (m, 2H, H2), 1649, sym(C_N) = 1537, asym(SO3) = 1281 and 1241, sym(CF3) =
8.64 (d, 1H, NH, 3JHH 7.9 Hz), 8.20 (m, 2H, H3), 7.27 (m, 5H, CH2Ph), 1220, asym(CF3) = 1157, sym(SO3) = 1025. 1H NMR (400 MHz,
4.95 (m, 1H, NHCH), 3.72 (s, 3H, COOCH3), A = 3.32 B = 3.15 (2H, sys- (CD3)2CO) : 8.93 (m, 2H, H2), 8.42 (d, NH, 3JHH 7.3 Hz), 8.01 (m, 2H,
tem ABX, CH2Ph, JAX 5.4 Hz, JBX 9.4 Hz, JAB 13.9 Hz). 13C{1H}NMR H3), 7.57 (m, 15H, HPh), 4.66 (qd, 1H, NHCH, 3JHH 7.3 Hz, 3JHH 7.3 Hz),
(400 MHz, (CD3)2CO) : 172.91 (s, 1C, COOCH3), 164.67 (s, 1C, CONH), 3.71 (s, 3H, COOCH3), 1.50 (d, 3H, CHCH3, 3JHH 7.4 Hz). 19F NMR
153.14 (s, 2C, C2), 148.62 (s, 1C, C4), 138.78 (s, 1C, CipsoCH2Ph), 131.06 (400 MHz, (CD3)2CO) : 79.91 (s, 1F, OTf). 31P{1H}NMR (400 MHz,
(s, 2C, CorthoCH2Ph), 130.30 (s, 1C, CparaCH2Ph), 128.68 (s, 2C, C3), 127.73 (CD3)2CO) : 17.0 and 12.74 (2 s br, 1P, AgPPh3). 31P{1H}NMR
(s, 2C, CmetaCH2Ph), 56.52 (s, 1C, NHCH), 53.62 (s, 1C, COOCH3), 38.88 (s, (300 MHz, (CD3)2CO, 203 K) : 12.5 (2d, 1P, AgPPh3, J109AgP 732.8 Hz,
1C, CH2Ph). UVvis, max nm: 232, 265 (br). J107AgP 635.0 Hz), 9.7 (2d, 2P, Ag(PPh3)2, J109AgP 507.3 Hz, J107AgP
439.6 Hz). 13C{1H}NMR (300 MHz, (CD3)2CO) : 174.36 (s, 1C,
COOCH3), 166.01 (s, 1C, CONH), 153.39 (s, 2C, C2), 144.59 (s, 1C, C4),
4.8. Synthesis of [Ag(L)(PPh3)n](OTf) (L1, n = 1 (10), n = 2 (11); L2, n = 1 135.75 (d, 6C, CorthoPh, 2JCP 15.5 Hz), 133.02 (s, 3C, CparaPh), 132.40
(13), n = 2 (14); L3, n = 1 (16), n = 2 (17)) (s br, 3C, CipsoPh), 131.17 (d, 6C, CmetaPh, 3JCP 8.8 Hz), 124.23 (s, 2C, C3),
53.42 (s, 1C, NHCH), 50.61 (s, 1C, COOCH3), 18.31 (s, 1C, CHCH3). UV
To a DMC solution (25 mL) of L1 (47 mg, 0.2 mmol), or L2 (42 mg, vis, max nm: 248, 275.
0.2 mmol), or L3 (57 mg, 0.2 mmol) was added 0.104 g (0.2 mmol) of
[Ag(OTf)PPh3], or 0.156 g (0.2 mmol) of [Ag(OTf)(PPh3)2] at room tem- 4.8.4. [Ag(L2)(PPh3)2](OTf) (14)
perature. After the addition, the reaction mixture was stirred for 1 h. The Yield: 180 mg, 91%. Anal. Calcd. (%) for C47H42AgF3N2O6P2S
volume was reduced to 5 mL, and addition of hexane afforded white (989.72): C, 57.04; H, 4.28; N, 2.83; S, 3.24. Found: C, 57.17; H, 4.22;
solids of 10, 11, 13, 14, 16 or 17. N, 2.85; S, 3.24. IR (cm 1): (NH) = 3312, (COcarboxylate) = 1740,
(COamide) = 1665, sym(C_N) = 1538, asym(SO3) = 1284 and
4.8.1. [Ag(L1)(PPh3)](OTf) (10) 1242, sym(CF3) = 1220, asym(CF3) = 1152, sym(SO3) = 1026. 1H
Yield: 127 mg, 84%. Anal. Calcd. (%) for C31H31AgF3N2O6PS (755.49): NMR (400 MHz, (CD3)2CO) : 8.77 (m, 2H, H2), 8.29 (s br, 1H, NH),
C, 49.28, H, 4.14; N, 3.71; S, 4.24. Found: C, 49.71; H, 3.88; N, 3.76; S, 7.84 (m, 2H, H3), 7.53 (m, 30H, HPh), 4.66 (qd, 1H, NHCH, 3JHH 7.3 Hz,
3
4.63. IR (cm1): (NH) = 3314, (COcarboxylate) = 1738, (COamide) = JHH 7.3 Hz), 3.71 (s, 3H, COOCH3), 1.49 (d, 3H, CHCH3, 3JHH 7.3 Hz).
19
1650, sym(C_N) = 1532, asym(SO3) = 1273 and 1240, sym(CF3) = F NMR (400 MHz, (CD3)2CO) : 80.03 (s, 1F, OTf). 31P{1H}NMR
1220, asym(CF3) = 1154, sym(SO3) = 1024. 1H NMR (400 MHz, (400 MHz, (CD3)2CO) : 12.6 (s br, 1P, AgPPh3). 31P{1H}NMR
(CD3)2CO) : 8.94 (m, 2H, H2), 8.18 (d, 1H, NH, 3JHH 8.2 Hz), 8.01 (m, (400 MHz, (CD3)2CO), 203 K) : 12.5 (2d, 1P, AgPPh3, J109AgP
2H, H3), 7.59 (m, 15H, HPh), 4.58 (dd, 1H, NHCH, 3JHH 6.4 Hz, 3JHH 726.6 Hz, J107AgP 629.2 Hz), 9.7 (2d, 2P, Ag(PPh3)2, J109AgP 500.6 Hz,
8.3 Hz), 3.74 (s, 3H, COOCH3), 2.28 (m, 1H, CH(CH3)2), 1.03 (m, 6H, J107AgP 317.7 Hz), 8.8 (2d, 3P, Ag(PPh3)3, J109AgP 433.7 Hz, J107AgP
CH(CH3)2). 19F NMR (400 MHz, (CD3)2CO) : 79.93 (s, 1F, OTf). 369.9 Hz). 13C{1H}NMR (400 MHz, (CD3)2CO) : 174.43 (s, 1C,
31 1
P{ H}NMR (400 MHz, (CD3)2CO) : 17.4 and 12.9 (2 s br, 1P, COOCH3), 166.30 (s, 1C, CONH), 152.76 (s, 2C, C2), 143.84 (s, 1C, C4),
AgPPh3). 31P{1H}NMR (300 MHz, (CD3)2CO, 203 K), 12.4 (2 d, 1P, 135.67 (d, 12C, CorthoPh, 2JCP 15.7 Hz), 132.02 (s, 6C, CparaPh), 132.61
AgPPh3, J109AgP 735.7 Hz, J107AgP 637.4 Hz), 9.6 (2 d, 2P, Ag(PPh3)2, (s br, 6C, CipsoPh), 131.15 (d, 12C, CmetaPh, 3JCP 9.2 Hz), 123.70 (s, 2C,
J109AgP 512.1 Hz, J107AgP 443.3 Hz). 13C{1H}NMR (400 MHz, C3), 53.40 (s, 1C, NHCH), 50.55 (s, 1C, COOCH3), 18.36 (s, 1C, CHCH3).
(CD3)2CO) : 172.48 (s, 1C, COOCH3), 165.69 (s, 1C, CONH), 152.67 (s,
2C, C2), 144.22 (s, 1C, C4), 134.83 (d, 6C, CorthoPh, 2JCP 16.1 Hz), 132.13 4.8.5. [Ag(L3)(PPh3)](OTf) (16)
(s, 3C, CparaPh), 131.64 (d, 3C, CipsoPh, 1JCP 30.1 Hz), 130.24 (d, 6C, CmetaPh, Yield: 159 mg, 99%. Anal. Calcd. (%) for C35H31AgF3N2O6PS (803.53):
3
JCP 9.9 Hz), 123.66 (s, 2C, C3), 59.49 (s, 1C, NHCH), 52.29 (s, 1C, C, 52.32; H, 3.89; N, 3.49; S, 3.99. Found: C, 52.43; H, 3.99; N, 3.80; S,
COOCH3), 31.41 (s, 1C, CH(CH3)2), 19.50 (s, 1C, CH(CH3)2), 18.86 (s, 4.25. IR (cm1): (NH) = 3299, (COcarboxylate) = 1739, (COamide) =
1C, CH(CH3)2). UVvis, max nm: 247, 275 (br). 1654, sym(C = N) = 1536, asym(SO3) = 1283 and 1241, sym(CF3) =
1220, asym(CF3) = 1155, sym(SO3) = 1025. 1H NMR (400 MHz,
(CD3)2CO) : 8.87 (s br, 2H, H2), 8.39 (d, 1H, NH, 3JHH 7.9 Hz), 7.88
4.8.2. [Ag(L1)(PPh3)2](OTf) (11) (s br, 2 H, H3), 7.65 (m, 15H, HPh), 7.56 (m, 5H, CH2Ph), 4.93 (m, 1H,
Yield: 173 mg, 85%. Anal. Calcd. (%) for C49H46AgF3N2O6P2S NHCH), 3.70 (s, 3H, COOCH3), A = 3.30 B = 3.15 (2H, system ABX,
(1017.78): C, 57.82; H, 4.56; N, 2.75; S, 3.15. Found: C, 57.43; H, 4.30; CH2Ph, JAX 5.4 Hz, JBX 9.4 Hz, JAB 13.9 Hz). 31P{1H}NMR (400 MHz,
N, 2.85; S, 3.52. IR (cm1): (NH) = 3301, (COcarboxylate) = 1739, (CD3)2CO) : 12.4 (s br, 1P, AgPPh3). 31P{1H}NMR (300 MHz,
(COamide) = 1664, sym(C_N) = 1532, asym(SO3) = 1282 and (CD3)2CO), 203 K) : 12.7 (2d, 1P, AgPPh3, J109AgP 754.0 Hz, J107AgP
1242, sym(CF3) = 1221, asym(CF3) = 1152, sym(SO3) = 1027. 1H 653.0 Hz), 9.8 (2d, 2P, Ag(PPh3)2, J109AgP 539.9 Hz, J107AgP 468.12 Hz).
13 1
NMR (300 MHz, (CD3)2CO) : 8.76 (m, 2H, H2), 8.03 (m, 1H, NH), 7.85 C{ H}NMR (400 MHz, (CD3)2CO) : 173.31 (s, 1C, COOCH3), 166.21
(m, 2H, H3), 7.54 (m, 30H, HPh), 4.58 (dd, 1H, NHCH, 3JHH 8.4 Hz, 3JHH (s, 1C, CONH), 153.40 (s, 2C, C2), 144.54 (s, 1C, C4), 139.17 (s, 1C,
6.4 Hz), 3.73 (s, 3H, COOCH3), 2.27 (m, 1H, CH(CH3)2), 1.02 (m, 6H, CipsoCH2Ph), 135.74 (d, 6C, CorthoPh, 2JCP 14.9 Hz), 133.02 (s, 3C, CparaPh),
CH(CH3)2). 19F NMR (400 MHz, (CD3)2CO) : 79.93 (s, 1F, OTf). 132.61 (d, 3C, CipsoPh, 1JCP 33.9 Hz), 131.17 (d, 6C, CmetaPh, 3JCP 7.5 Hz),
31 1
P{ H}NMR (300 MHz, (CD3)2CO) : 11.5 (s br, 2P, AgPPh3). 131.04 (s, 2C, CorthoCH2Ph), 130.22 (s, 1C, CparaCH2Ph), 128.55 (s, 2C,
31 1
P{ H}NMR (300 MHz, (CD3)2CO, 203 K), 12.3 (2d, 1P, AgPPh3, J109Ag CmetaCH2Ph), 124.16 (s br, 2C, C3), 56.38 (s, 1C, NHCH), 53.49 (s, 1C,
P 716.9 Hz, J107AgP 622.1 Hz), 9.5 (2d, 1P, Ag(PPh3)2, J109AgP 492.7 Hz, COOCH3), 38.77 (s, 1C, CH2Ph). UVvis, max nm: 248, 275.
J107AgP 426.9 Hz, 8.8 (2d, 3P, Ag(PPh3)3, J107 AgP 367.1 Hz).
13
C{1H}NMR (300 MHz, (CD3)2CO) : 152.61 (s, 2C, C2), 135.70 (d, 4.8.6. [Ag(L3)(PPh3)2](OTf) (17)
12C, CorthoPh, 2JCP 15.4 Hz,), 132.13 (s, 6C, CparaPh), 132.97 (d, 12C, CmetaPh, Yield: 204 mg, 96%. Anal. Calcd. (%) for C53H46AgF3N2O6P2S
3
JCP 8.8 Hz), 123.65 (s, 2C, C3), 60.30 (s, 1C, NHCH), 53.20 (s, 1C, (1065.82): C, 59.73; H, 4.35; N, 2.63; S, 3.01. Found: C, 59.45; H, 4.36;
COOCH3), 32.41 (s, 1C, CH(CH3)2), 20.47 (s, 1C, CH(CH3)2), 19.80 (s, N, 2.93; S, 3.29. IR (cm 1): (NH) = 3313, (COcarboxylate) = 1741,
1C, CH(CH3)2). (COamide) = 1667, sym(C_N) = 1539, asym(SO3) = 1282 and
142 L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144

1243, sym(CF3) = 1221, asym(CF3) = 1152, sym(SO3) = 1027. 1H (s, 1C, CONH), 157.91 (d, 1C, C2, 1JCP 53.71 Hz), 155.20 (s br, 1C, C6),
NMR (400 MHz, (CD3)2CO) : 8.74 (m, 2H, H2), 8.29 (s br, 1H, NH), 151.59 (s, 2C, C2), 144.77 (s, 1C, C4), 141.53 (s, 1C, C4), 135.56 (s br,
8.03 (m, 2H, H3), 7.39 (m, 30H, HPh), 7.28 (m, 5H, CH2Ph), 4.90 (m, 1H, 4C, CorthoPh), 133.49 (s, 2C, CparaPh), 132.47 (m, 2C, C3), 131.49 (s br, 4C,
NHCH), 3.70 (s, 3H, COOCH3), A = 3.30 B = 3.15 (2H, system ABX, CmetaPh), 128.45 (s, 1C, C5), 124.15 (s, 2C, C3), 53.42 (s, 1C, NHCH),
CH2Ph, JAX 5.4 Hz, JBX 9.4 Hz, JAB 13.9 Hz). 31P{1H}NMR (400 MHz, 50.61 (s, 1C, COOCH3), 18.28 (s, 1C, CHCH3). UVvis, max nm: 234, 264.
(CD3)2CO) : 12.3 (s br, 2P, Ag(PPh3)2). 31P{1H}NMR (300 MHz,
(CD3)2CO), 203 K) : 12.6 (2d, 1P, AgPPh3, J109AgP 737.3 Hz, J107AgP 4.9.3. [Ag(L3)(PPh2py)](OTf) (18)
638.4 Hz), 9.9 (2d, 2P, Ag(PPh3)2, J109AgP 513.7 Hz, J107AgP 445.0 Hz). Yield: 157 mg, 97%. Anal. Calcd. (%) for C34H30AgF3N3O6PS (804.52):
13
C{1H}NMR (400 MHz, (CD3)2CO) : 173.37 (s, 1C, COOCH3), 166.47 C, 50.76; H, 3.76; N, 5.22; S, 3.99. Found: C, 50.83; H, 3.84; N, 5.65; S,
(s, 1C, CONH), 152.85 (s, 2C, C2), 143.81 (s, 1C, C4), 139.20 (s, 1C, 4.04. IR (cm1): (NH) = 3322, (COcarboxylate) = 1740, (COamide) =
CipsoCH2Ph), 135.67 (d, 12C, CorthoPh, 2JCP 15.7 Hz), 132.90 (s, 6C, CparaPh), 1666, sym(C_N) = 1540, asym(SO3) = 1265 and 1250, sym(CF3) =
132.62 (s br, 6C, CipsoPh), 131.14 (d, 12C, CmetaPh, 3JCP 9.0 Hz), 131.04 (s, 1221, asym(CF3) = 1151, sym(SO3) = 1028. 1H NMR (400 MHz,
2C, CorthoCH2Ph), 130.22 (s, 1C, CparaCH2Ph), 128.54 (s, 2C, CmetaCH2Ph), (CD3)2CO) : 9.14 (s br, 1H, H6), 8.84 (m, 2H, H2), 8.35 (m, 1H, NH),
123.67 (s br, 2C, C3), 56.31 (s, 1C, NHCH), 53.46 (s, 1C, COOCH3), 8.17 (m, 1H, H4), 7.89 (m, 3H, H3, 5), 7.63 (m, 11H, HPh,3), 7.28 (m,
38.80(s, 2C, CH2Ph). 5H, CH2Ph), 4.93 (m, 1H, NHCH), 3.72 (s, 3H, COOCH3), A = 3.31
B = 3.16 (2H, system ABX, CH2Ph, JAX 5.4 Hz, JBX 9.4 Hz, JAB 13.9 Hz).
4.9. Synthesis of [Ag(L)(PPh2py)](OTf) (L1 (12), L2 (15), L3 (18)) 31 1
P{ H}NMR (400 MHz, (CD3)2CO) : 30.1 (s br, 1P, AgPPh2py).
31 1
P{ H}NMR (300 MHz, (CD3)2CO), 203 K) : 17.9 (2 m, 1P, AgPPh2py,
To a DMC solution (25 mL) of L1 (47 mg, 0.2 mmol), L2 (42 mg, Javerage 623.4 Hz). 13C{1H}NMR (400 MHz, (CD3)2CO) : 173.36 (s, 1C,
0.2 mmol), or L3 (57 mg, 0.2 mmol) was added [Ag(OTf)(PPh2py)] COOCH3), 166.31 (s, 1C, CONH), 155.37 (s br, 1C, C6), 151.56 (s, 2C,
(105 mg, 0.2 mmol) at room temperature. After the addition, the reac- C2), 144.56 (s, 1C, C4), 141.65 (s, 1C, C4), 139.17 (s, 1C, CipsoCH2Ph),
tion mixture was stirred for 1 h. The volume was reduced to 5 mL, and 135.57 (s br, 4C, CorthoPh), 133.54 (s br, 2C, CparaPh), 132.55 (s br, 1C,
addition of hexane afforded compounds 12, 15 and 18. C3), 131.54 (s br, 4C, CmetaPh), 131.05 (s, 2C, CorthoCH2Ph), 130.25 (s, 1C,
CparaCH2Ph), 128.57 (s, 2C, CmetaCH2Ph), 123.91 (s, 2C, C3), 56.32 (s, 1C,
NHCH), 53.49 (s, 1C, COOCH3), 38.80 (s, 1C, CH2Ph). UVvis, max nm:
230, 264.

4.10. Synthesis of [Cu(L)(PPh3)]NO3 (L1 (19), L2 (20), L3 (21))

These reactions were carried out under Ar, in dried solvents and
4.9.1. [Ag(L1)(PPh2py)](OTf) (12)
degassed prior to use. To a DMC solution (25 mL) was added L1
Yield: 124 mg, 82%. Anal. Calcd. (%) for C30H30AgF3N3O2PS (756.48):
(47 mg, 0.2 mmol), L2 (42 mg, 0.2 mmol), or L3 (57 mg, 0.2 mmol)
C, 47.63; H, 4.00; N, 5.55; S, 4.24. Found: C, 47.55; H, 3.90; N, 5.43; S,
and [Cu(NO3)(PPh3)2] (130 mg, 0.2 mmol), at room temperature.
3.94. IR (cm1): (NH) = 3287, (COcarboxylate) = 1736, (COamide) =
After the addition, the reaction mixture was stirred for 1 h and then
1662, sym(C_N) = 1534, asym(SO3) = 1277 and 1242, sym(CF3) =
was concentrated under vacuum. The addition of hexane gave yellow
1221, asym(CF3) = 1153, sym(SO3) = 1027. 1H NMR (400 MHz,
precipitates that were ltered off.
(CD3)2CO) : 9.27 (s br, 1H, H6), 8.69 (m, 2H, H2), 8.22 (d, 1H, NH,
3
JHH 7.9 Hz), 8.14 (m, 1H, H4), 7.89 (m, 2H, H3), 7.80 (m, 1H, H5), 7.40
(m, 11H, HPh,3), 4.54 (m, 1H, NHCH), 3.72 (s, 3H, COOCH3), 2.27 (m, 4.10.1. [Cu(L1)(PPh3)](NO3) (19)
1H, CH(CH3)2), 1.02 (m, 6H, CH(CH3)2). 19F NMR (400 MHz, Yield: 97 mg, 78%. Anal. Calcd. (%) for C30H31CuN3O6P (624.10): C,
(CD3)2CO) : 80.07 (s, 1F, OTf). 31P{1H}NMR (400 MHz, (CD3)2CO) 57.73; H, 5.01; N, 6.73. Found: C, 57.69; H, 5.46; N, 6.83. IR (cm 1):
: 18.7 (s br, 1P, AgPPh2py). 31P{1H}NMR (300 MHz, (CD3)2CO, 203 K) (NH) = 3203, (COcarboxylate) = 1732, (COamide) = 1662,
: 20.0 and 15.0 (2 m, 1P, AgPPh2py, Javerage 666.5Hz). 13C{1H}NMR sym(C_N) = 1537, (NO3) = 1095, 1022, 1009, 828, 692. 1H NMR
(400 MHz, (CD3)2CO) : 173.42 (s, 1C, COOCH3), 166.78 (s, 1C, CONH), (400 MHz, (CD3)2CO) : 8.71 (m, 1H, H2), 7.99 (s br, 1H, NH), 7.79 (m,
157.93 (d, 1C, C2, 1JCP 55.8 Hz), 155.20 (d, 1C, C6, 3JCP 16.0 Hz), 151.59 2H, H3), 7.42 (m, 15H, HPh), 4.57 (dd, 1H, NHCH, 3JHH 4.9, 3JHH 8.5 Hz),
(s, 2C, C2), 144.94 (s, 1C, C4), 141.43 (s, 1C, C4), 135.48 (d, 4C, CorthoPh, 3.72 (s, 3H, COOCH3), 2.26 (m, 1H, CH(CH3)2), 1.02 (m, 6H, CH(CH3)2).
31 1
2
JCP 14.2 Hz), 133.40 (s, 2C, CparaPh), 132.35 (m, 1C, H3), 131.46 (d, 4C, P{ H}NMR (400 MHz, (CD3)2CO) : 0.0 (s, CuPPh3). 13C{1H}NMR
CmetaPh, 3JCP 8.2 Hz), 130.48 (m, 2C, CipsoPh), 128.38 (s, 1C, C5), 124.32 (300 MHz, CDCl3) : 172.19 (s, 1C, COOCH3), 165.31 (s, 1C, CONH),
(s, 2C, C3), 60.45 (s, 1C, NHCH), 53.21 (s, 1C, COOCH3), 32.21 (s, 1C, 150.63 (s, 2C, C2), 133.60 (d, 6C, CorthoPh, 2JCP 13.0 Hz), 131.73 (d, 3C,
CH(CH3)2), 20.43 (s, 1C, CH(CH3)2), 19.84 (s, 1C, CH(CH3)2). UVvis, CipsoPh, 1JCP 32.9 Hz), 130.18 (s, 3C, CparaPh), 128.82 (d, 6C, CmetaPh, 3JCP
max nm: 230, 264. 5.6 Hz), 57.59 (s, 1C, NHCH), 52.42 (s, 1C, COOCH3), 31.56 (s, 1C,
CH(CH3)2), 18.94 (s, 1C, CH(CH3)2), 17.93 (s, 1C, CH(CH3)2).
4.9.2. [Ag(L2)(PPh2py)](OTf) (15)
Yield: 128 mg, 88%. Anal. Calcd. (%) for C28H26AgF3N3O6PS (728.43): 4.10.2. [Cu(L2)(PPh3)](NO3) (20)
C, 46.17; H, 3.60; N, 5.77; S, 4.40. Found: C, 46.42; H, 3.74; N, 5.61; S, Yield: 82 mg, 69%. Anal. Calcd. (%) for C28H27CuN3O6P (596.05): C,
4.04. IR (cm1): (NH) = 3316, (COcarboxylate) = 1740, (COamide) = 56.42; H, 4.57; N, 7.05. Found: C, 56.65; H, 4.61; N, 4.79. IR (cm 1):
1663, sym(C_N) = 1539, asym(SO3) = 1275 and 1243, sym(CF3) = (NH) = 3296, (COcarboxylate) = 1741, (COamide) = 1665,
1221, asym(CF3) = 1154, sym(SO3) = 1027. 1H NMR (400 MHz, sym(C_N) = 1540, (NO3) = 1095, 1024, 1024, 849, 692. 1H NMR
(CD3)2CO) : 9.25 (s br, 1H, H6), 8.73 (m, 2H, H2), 8.34 (m, 1H, NH), (400 MHz, CDCl3) : 8.55 (m, 2H, H2), 7.66 (m, 2H, H3), 7.43 (d, 1H,
8.16 (m, 1H, H4), 7.89 (m, 2H, H3), 7.81 (m, 1H, H5), 7.46 (m, 11H, NH, 3JHH 7.0 Hz), 7.27 (m, 15H, HPh), 4.73 (qd, 1H, NHCH, 3JHH 7.2 Hz,
HPh,3), 4.64 (qd, 1H, NHCH, 3JHH 7.3 Hz, 3JHH 7.3 Hz), 3.70 (s, 3H, 3
JHH 7.2 Hz), 3.77 (s, 3H, COOCH3), 1.51 (d, 3H, CHCH3, 3JHH 7.2 Hz).
COOCH3), 1.49 (d, 3H, CHCH3, 3JHH 7.3 Hz). 19F NMR (400 MHz, 31 1
P{ H}NMR (400 MHz, CDCl3) : 0.0 (s, 1P, CuPPh3). 13C{1H}NMR
(CD3)2CO) : 80.06 (s, 1F, OTf). 31P{1H}NMR (400 MHz, (CD3)2CO) (400 MHz, CDCl3) : 174.15 (s, 1C, COOCH3), 164.83 (s, 1C, CONH),
: 20.4 and 14.4 (s br, 1P, AgPPh2py). 31P{1H}NMR (400 MHz, 135.58 (d, 6C, CorthoPh, 2JCP 13.9 Hz), 132.61 (d, 3C, CipsoPh, 1JCP 33.8 Hz),
(CD3)2CO), 203 K) : 17.4 (2 m, AgPPh2py, Javerage 695.8 Hz). 130.17 (s, 3C, CparaPh), 128.82 (d, 6C, CmetaPh, 3JCP 7.5 Hz), 52.70 (s, 1C,
13
C{1H}NMR (400 MHz, (CD3)2CO) : 174.36 (s, 1C, COOCH3), 166.01 NHCH), 48.70 (s, 1C, COOCH3), 18.25 (s, 1C, CHCH3).
L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144 143

4.10.3. [Cu(L3)(PPh3)](NO3) (21) Munchen, Germany). The IC50 was calculated by nonlinear regression
Yield: 100 mg, 76%. Anal. Calcd. (%) for C34H31CuN3O6P (672.16): C, analysis.
60.76; H, 4.65; N, 6.25. Found: C, 60.43; H, 4.16; N, 6.71. IR (cm 1):
(NH) = 3246, (COcarboxylate) = 1742, (COamide) = 1666, 4.14. Flow cytometry analysis of cell death
sym(C_N) = 1539, (NO3) = 1094, 1023, 1023, 844, 692. 1H NMR
(400 MHz, DMSO) : 9.25 (m, 2H, H2), 8.82 (s br, 1H, NH), 7.77 (s br, NIH-3 T3 cells were seeded in 6-well plate format at 100,000 cells/
2H, H3), 7.40 (m, 20H, CH2Ph, HPh), 4.75 (m, 1H, NHCH), 3.71 (s, 3H, well using 1 mL DMEM media supplemented (see Table 3). Cells were
COOCH3), A = 3.25 B = 3.14 (2H, system ABX, CH2Ph, JAB 5.2 Hz, JBX exposed for 24 h to IC50 concentrations of complexes and ligands.
10.2 Hz, JAX 13.7 Hz). 31P{1H}NMR (400 MHz, DMSO) : 3.7 (s, 1P, Cells from each individual well were centrifuged 400 x g for 5 min,
CuPPh3). 13C{1H}NMR (300 MHz, (CD3)2CO) : 173.46 (s, 1C, COOCH3), washed two times with cold PBS and then resuspended in 200 L of
166.86 (s br, 1C, CONH), 152.32 (s br, 2C, C2), 139.17 (s, 1C, CipsoCH2Ph), binding buffer. 100 L of each cell solution was transferred to a culture
135.41 (d, 6C, CorthoPh, 2JCP 14.9 Hz), 134.17 (d, 3C, CipsoPh, 1JCP 32.0 Hz), tube and added 5 L of Annexin V-FITC and 5 L of PI (stock solution
132.22 (s, 3C, CparaPh), 131.04 (s, 2C, CorthoCH2Ph), 130.80 (d, 6C, CmetaPh, 50 g/mL). After incubation for 20 min in the dark, cell suspensions
3
JCP 9.3 Hz), 130.24 (s, 1C, CparaCH2Ph), 128.57 (s, 2C, CmetaCH2Ph), 56.23 were added to 400 L of binding buffer, gently homogenized, and ana-
(s, 1C, NHCH), 53.45 (s, 1C, COOCH3), 38.88 (s, 1C, CH2Ph). lyzed by ow cytometry (Coulter Epics XL-MCL, Coulter, Krefeld,
Germany).
4.11. Crystal structure determination
Abbreviations
Data were registered on a Bruker Smart APEX CCD (7) or Xcalibur
Oxford Diffraction (8) diffractometers. The crystals were mounted in asym asymmetry
inert oil on glass bers and transferred to the cold gas stream of the CDCl3 chloroform-d
diffractometers. Data were collected using monochromated Mo K ra- (CD3)2CO acetone-d6
diation ( = 0.710 73) in scans. Absorption corrections based on mul- COSY correlation spectroscopy
tiple scans were applied with the program SADABS [61]. The structures DMC dichlorometane
were solved by direct methods and rened on F2 using the program DMEM dulbecco's modied eagle's medium
SHELXL-97 [62]. All non-hydrogen atoms were rened anisotropically. HSQC heteronuclear single quantum coherence spectroscopy
Hydrogen atoms were included by using a riding model. CCDC- Hz Hertz
1,423,474 (7) and CCDC-1,423,475 (8) contain the supplementary crys- FCS fetal calf serum
tallographic data for this paper. These data can be obtained free of FITC uorescein isothiocyanate
charge from The Cambridge Crystallographic Data Centre via www. IC50 half maximal inhibitory concentration
ccdc.cam.ac.uk/data_request/cif. IR infrared
MTT 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium
bromide
4.12. Cell cultures
NCS newborn calf serum
NMR nuclear magnetic resonance
Cell lines were cultivated in Dulbecco's Modied Eagle's medium
OTf triuoromethanesulfonate
(DMEM, D7777 Sigma-Aldrich, Steinheim, Germany) supplemented
P penicillin
with the according serum (FCS, fetal calf serum or NCS, newborn calf
PBS phosphate buffered saline
serum) (Table 3) and 1% antibiotics (penicillin/100 U/mL and strepto-
PI propidium iodide
mycin/100 g/mL) in a humidied atmosphere of 95% air/37 C/5%
PS phosphatidylserine
CO2. After 34 days the cells had grown to conuence and were then de-
Py pyridine
tached with trypsin and cultured in a new cell culture ask.
ROS reactive oxygen species
S streptomycin
4.13. Cytotoxicity assays by MTT sym symmetry

The MTT assay was used to determine cell viability as an indicator for Acknowledgments
cell sensitivity to the complexes (MTT, Sigma-Aldrich, Steinheim,
Germany). The authors thank the Ministerio de Economa y Competitividad-
Exponentially growing cells were seeded with a dened number of FEDER CTQ2013-48635-C2-1-P and DGA-FSE (E77) for nancial
cells (see Table 3), in a 96-well at-bottomed microplate and 48 h support.
later they were incubated with the compounds. The complexes were
dissolved in DMSO and tested in concentrations ranging from 1 to
Appendix A. Supplementary data
50 M. Cytotoxicity of tested compounds was evaluated by the MTT
method [63]. Controls were made by incubating cells with supplement-
Supplementary data to this article can be found online at http://dx.
ed media and supplemented media with 5% DMSO. Optical density was
doi.org/10.1016/j.jinorgbio.2015.12.018.
measured at 570 nm using a 96-well multiscanner autoreader (Bio-Rad,

References
Table 3
[1] E.R.T. Tiekink, Crit. Rev. Oncol. Hematol. 42 (2002) 225248.
DMEM additives for each cell line and the cell number per well. [2] I. Ott, Coord. Chem. Rev. 153 (2009) 16701681.
[3] C.M. Che, R.W.Y. Sun, Chem. Commun. 47 (2011) 95549560.
Cell line DMEM additives Cell number per well
[4] B. Bertrand, A. Casini, Dalton Trans. 43 (2014) 42094219.
Hep-G2 10% FCS, 1% P/S 10000 [5] C.K. Mirabelli, R.K. Johnson, D.T. Hill, L.F. Faucette, G.R. Girard, G.Y. Kuo, C.M. Sung,
A-549 10% FCS, 1% P/S 8000 S.T. Crooke, J. Med. Chem. 29 (1986) 218223.
HeLa 10% FCS, 1% P/S 8000 [6] S.J. Berners-Price, G.R. Girard, D.T. Hill, B.M. Sutton, P.S. Jarrett, L.F. Faucette, R.K.
NIH-3 T3 10% NCS, 1% P/S 6000 Johnson, C.K. Mirabelli, P.J. Sadler, J. Med. Chem. 33 (1990) 13861392.
[7] J.J. Liu, P. Galettis, A. Farr, L. Maharaj, H. Samarasinha, A.C. McGechan, B.C. Baguley,
MCF-7 10% FCS, 1% P/S 5000
R.J. Bowen, S.J. Berners-Price, M.J. McKeage, J. Inorg. Biochem. 102 (2008) 303310.
144 L. Ortego et al. / Journal of Inorganic Biochemistry 156 (2016) 133144

[8] E. Vergara, A. Casini, F. Sorrentino, O. Zava, E. Cerrada, M.P. Rigobello, A. Bindoli, M. [34] S. Patil, A. Deally, B. Gleeson, H. Muller-Bunz, F. Paradisi, M. Tacke, Metallomics 3
Laguna, P.J. Dyson, Chem. Med. Chem. 5 (2010) 96102. (2011) 7488.
[9] L. Kaps, B. Biersack, H. Mller-Bunz, K. Mahal, J. Mnzner, M. Tacke, T. Mueller, R. [35] C. Santini, M. Pellei, V. Gandin, F. Tisato, C. Marzano, Chem. Rev. 114 (2014)
Schobert, J. Inorg. Biochem. 106 (2012) 5258. 815862.
[10] G. Marcon, S. Carotti, M. Coronnello, L. Messori, E. Mini, P. Orioli, T. Mazzei, M.A. [36] C. Marzano, V. Gandin, M. Pellei, D. Colavito, G. Papini, G. Gioia Lobbia, E. Del Giudice,
Cinellu, G. Minguetti, J. Med. Chem. 45 (2002) 16721677. M. Porchia, F. Tisato, C. Santini, J. Med. Chem. 51 (2008) 798808.
[11] C.M. Che, R.W.Y. Sun, W.Y. Yu, C.B. Ko, N. Zhu, H. Sun, Chem. Commun. (2003) [37] C. Santini, M. Pellei, G. Papini, B. Morresi, R. Galassi, S. Ricci, F. Tisato, M. Porchia, M.P.
17181719. Rigobello, V. Gandin, C. Marzano, J. Inorg. Biochem. 105 (2011) 232240.
[12] L. Ronconi, C. Marzano, P. Zanello, M. Corsini, G. Miolo, C. Macca, A. Trevisan, D. [38] M.S. Balakrishna, D. Suresh, A. Rai, J.T. Mague, D. Panda, Inorg. Chem. 49 (2010)
Fregona, J. Med. Chem. 49 (2006) 16481657. 87908801.
[13] J.S. Casas, M.V. Castao, M.C. Cifuentes, J.C. Garca-Monteagudo, A. Snchez, J. Sordo, [39] V. Gandin, M. Pellei, F. Tisato, M. Porchia, C. Santini, C. Marzano, J. Cell. Mol. Med. 16
U. Abram, J. Inorg. Biochem. 98 (2004) 10091016. (2012) 142151.
[14] R. Buckley, A. Elsome, S. Fricker, G. Henderson, B. Theobald, R. Parish, B. Howe, L. [40] K. Lazarou, B. Bednarz, M. Kubicki, I.I. Verginadis, K. Charalabopoulos, N.
Kelland, J. Med. Chem. 39 (1996) 52085214. Kourkoumelis, S.K. Hadjikakou, Inorg. Chim. Acta 363 (2010) 763772.
[15] A. Bindoli, M.P. Rigobello, R. Scutari, C. Gabbiani, A. Casini, L. Messori, Coord. Chem. [41] R. Starosta, K. Stokowa, M. Florek, J. Krol, A. Chwilkowska, J. Kulbacka, J. Saczko, J.
Rev. 253 (2009) 16921707. Skala, M. Jezowska-Bojczuk, J. Inorg. Biochem. 105 (2011) 11021108.
[16] P.T. Barnard, S.J. Berners-Price, Coord. Chem. Rev. 251 (2007) 18891902. [42] M.-L. Teyssot, A.-S. Jarrousse, A. Chevry, A. De Haze, C. Beaudoin, M. Manin, S.P.
[17] S.J. Berners-Price, A. Filipovska, Metallomics 3 (2011) 863873. Nolan, S. Dez-Gonzlez, L. Morel, A. Gautier, Chem. Eur. J. 15 (2009) 314318.
[18] S. Urig, K. Fritz-Wolf, R. Reau, C. Herold-Mende, K. Toth, E. Davioud-Charvet, K. [43] V. Fernndez-Moreira, M.L. Ortego, C.F. Williams, M.P. Coogan, M.D. Villacampa, M.C.
Becker, Angew. Chem. Int. Ed. 45 (2006) 18811886. Gimeno, Organometallics 31 (2012) 59505957.
[19] J.L. Hickey, R.A. Ruhayel, P.J. Barnard, M.V. Baker, S.J. Berners-Price, A. Filipovska, J. [44] A.M. Blanco-Rodrguez, M. Towrie, J. Scora, S. Zlis, A. Vlcek Jr., Inorg. Chem. 50
Am. Chem. Soc. 130 (2008) 1257012571. (2011) 61226134.
[20] E. Schuh, C. Pger, A. Citta, A. Folda, M.P. Rigobello, A. Bindoli, A. Casini, F. Mohr, J. [45] M. Bardaj, O. Crespo, A. Laguna, A.K. Fischer, Inorg. Chim. Acta 304 (2000) 716.
Med. Chem. 55 (2012) 55185528. [46] K. Nomiya, K. Tsuda, Y. Tanabe, H. Nagano, J. Inorg. Biochem. 69 (1998) 914.
[21] A. Casini, C. Hartinger, C. Gabbiani, E. Mini, P.J. Dyson, B.K. Keppler, L. Messori, J. [47] L. Lettko, J.S. Wood, M.D. Rausch, Inorg. Chim. Acta 38 (2000) 3744.
Inorg. Biochem. 102 (2008) 564575. [48] L. Muertterties, C.W. Alegranti, J. Am. Chem. Soc. 92 (1970) 41144115.
[22] J. Lemke, A. Pinto, P. Niehoff, V. Vasylyeva, N. Metzler-Nolte, Dalton Trans. 35 (2009) [49] E.L. Muertterties, C.W. Alegranti, J. Am. Chem. Soc. 94 (1972) 63866391.
70637070. [50] S.J. Berners-Price, R.J. Bowen, P.J. Harvey, P.C. Healy, G.A. Koutsantonis, J. Chem. Soc.
[23] B.. Glii, U. Rychlewska, M.I. Djuran, Dalton Trans. 41 (2012) 68876901. Dalton Trans. (1998) 17431750.
[24] A. Gutierrez, J. Bernal, M.D. Villacampa, C. Cativiela, A. Laguna, M.C. Gimeno, Inorg. [51] O. Crespo, M.C. Gimeno, A. Laguna, C. Larraz, Z. Naturforsch. 64b (2009) 15251534.
Chem. 52 (2013) 64736480. [52] Q. Zhou, Y.-H. Fu, X.-B. Li, G.-Y. Chen, S.-Y. Wu, X.-P. Song, Y.-P. Liu, C.-R. Han,
[25] A. Gutirrez, L. Gracia-Fleta, I. Marzo, C. Cativiela, A. Laguna, M.C. Gimeno, Dalton Phytochem. Lett. 11 (2015) 296300.
Trans. 43 (2014) 1705417066. [53] J. Shao, Z.-Y. Ma, A. Li, Y.-H. Liu, C.-Z. Xie, Z.-Y. Qiang, J.-Y. Xu, J. Inorg. Biochem. 136
[26] C.N. Banti, S.K. Hadjikakou, Metallomics 5 (2013) 569596. (2014) 1323.
[27] J.S. Medici, M. Peana, V.M. Nurchi, I. Lachowicz, G. Crisponi, M.A. Zoroddu, Coord. [54] S. Osbild, L. Brault, E. Battaglia, D. Bagre, Anticancer Res. 26 (2006) 35953600.
Chem. Rev. 284 (2015) 329350. [55] G. Koopman, C.P.M. Reutelingsperger, G.A.M. Kuitjen, R.M.J. Keehnen, S.T. Pals, M.H.J.
[28] K.I. Batarseh, Curr. Med. Chem. 20 (2013) 23632373. van Oers, Blood 84 (1994) 14151420.
[29] V.T. Yilmaz, E. Gocmen, C. Icsel, M. Cengiz, S.Y. Susluer, O. Buyukgungor, J. [56] G. Zhang, V. Gurtu, S.R. Kain, G. Yan, BioThecniques 23 (1997) 525531.
Photochem. Photobiol. B Biol. 131 (2014) 3142. [57] H. Lecoeur, Exp. Cell Res. 277 (2002) 114.
[30] C. Pettinari, F. Marchetti, G. Lupidi, L. Quassinti, M. Bramucci, D. Petrelli, L.A. Vitali, [58] C.M. Henry, E. Hollville, S.J. Martin, Methods 61 (2013) 9097.
M.F. da Silva, L.M. Martins, P. Smolenski, A.J. Pombeiro, Inorg. Chem. 50 (2011) [59] R. Uson, A. Laguna, Inorg. Synth. 21 (1982) 7174.
1117311183. [60] N.W. Alcock, P. Moore, P.A. Lampe, K.F. Mok, J. Chem. Soc. Dalton Trans. (1982) 207.
[31] C.N. Banti, A.D. Giannoulis, N. Kourkoumelis, A.M. Owczarzak, M. Poyraz, M. Kubicki, [61] G.M. Sheldrick, SADABS, Program for Absorption Correction, University of Gttingen,
K. Charalabopoulos, S.K. Hadjikakou, Metallomics 4 (2012) 545560. Gttingen, Germany, 1996.
[32] F. Hackenberg, M. Tacke, Dalton Trans. 43 (2014) 81448153. [62] G.M. Sheldrick, SHELXL-97, Program for Crystal Structure Renement, University of
[33] M.L. Teyssot, A.S. Jarrousse, M. Manin, A. Chevry, S. Roche, F. Norre, C. Beau-Doin, L. Gttingen, Gttingen, Germany, 1997.
Morel, D. Boyer, R. Mahiou, A. Gautier, Dalton Trans. 35 (2009) 68946902. [63] T. Mossman, J. Immunol. Methods 65 (1983) 5563.

You might also like