Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Inorganica Chimica Acta xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Inorganica Chimica Acta


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

Design, spectral characterization, anti-tumor and anti-inflammatory


activity of triorganotin(IV) hydroxycarboxylates, apoptosis inducers:
In vitro assessment of induction of apoptosis by enzyme,
DNA-fragmentation, acridine orange and comet assays
Mala Nath a,⇑, Monika Vats a, Partha Roy b
a
Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, India
b
Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India

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

Article history: Interaction of triorganotin(IV) chlorides with sodium salt of hydroxycarboxylic acids results in the
Received 29 October 2013 formation of triorganotin(IV) hydroxycarboxylates, R3Sn(L) [R = Me, n-Bu and Ph; L = anion of glucuronic
Received in revised form 17 January 2014 (HGlu), gallic (HGal) and mandelic (HMal) acid]. They exhibit trigonal–bipyramidal geometry which is
Accepted 22 February 2014
well supported by elemental analysis, IR, 1H, 13C, 119Sn NMR and ESI-MS spectral data. Triorganotin(IV)
Available online xxxx
hydroxycarboxylates have been screened in vitro against five cancer cell lines of human origin viz.
Antitumor Active Organotin Compounds MCF-7, HEK-293, PC-3, HCT-15 and HepG-2. Title complexes are found to be cytotoxic to mildly cytotoxic,
and exhibited IC50 values in the range 4–30 lg/mL. The results of enzyme assays viz. glutathione reduc-
Keywords: tase, glutathione peroxidase, total glutathione content and lipid peroxidase assay on MCF-7 cells indicate
Anti-cancer activity that the reactive oxygen species generated in the cancer cells by triorganotin(IV) hydroxycarboxylates is
Apoptosis responsible for cell death. Marginal increase of lactate dehydrogenase suggests that necrosis is also
DNA cleavage occurring to a small extent. DNA (deoxyribonucleic acid) fragmentation assay, acridine orange assay
Anti-inflammatory activity and comet assay clearly support that the cell death is mainly due to apoptosis. The results obtained
Triorganotin(IV) hydroxycarboxylate for the in vivo anti-inflammatory activity (% inhibition) and toxicity (LD50 in mg/kg) suggested that the
complexes have low toxicity and good anti-inflammatory activity.
Ó 2014 Elsevier B.V. All rights reserved.

1. Introduction proliferation of cancer cells for which the replication of DNA is to


be arrested. A number of cancer chemotherapeutic drugs are
Globally cancer among the various deadly diseases is one of the commercially available for treatment of different types of cancers,
leading health concerns for humans, accounting for more than 15% but each of which has one or other severe side effects [2]. That is
of human deaths. Recently, it has been reported by WHO that why; one of the most rapidly developing areas of pharmaceutical
about 27 million new cancer cases and 11.5 million deaths due to research is the discovery, design and synthesis of robust, effective
cancer are expected in the next 2 decades [1]. Cancers, caused by and selective cancer chemotherapeutic drugs which should have
abnormal and uncontrolled cell division, are usually derived from minimum side effects.
numerous tissues with multiple etiologies and endless combina- Further, cancer chemotherapy based on metallotherapeutic
tion of genetic and/or epigenetic alterations; therapies for cancers drugs has gained momentum after the serendipitous discovery of
are as diverse as the disease itself. The treatments for cancer cis-platin (cis-diamminedichloroplatinum(II)) [3,4]. However,
include surgery, radiotherapy, photodynamic therapy and chemo- despite its remarkable success, there are well-known drawbacks
therapy, and chemotherapy is based on the inhibition of the rapid associated with Pt drugs, viz. nephrotoxicity or neurotoxicity,
intrinsic acquired drug resistance and patient compliance [5,6].
Abbreviations: Bz-d6, deteuratedbenzene; HGlu, glucuronic acid; HGal, gallic
Therefore, this discovery stimulated the search for other
acid; HMal, mandelic acid; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazo- metal-based chemotherapeutics with potential antitumor activity
lium bromide; DNA, deoxyribonucleic acid. having fewer side effects [7,8]. Among these, organotins have
⇑ Corresponding author. Tel.: +91 9897135529. emerged as potential biologically active metallopharmaceuticals,
E-mail addresses: malanfcy@iitr.ac.in, malanfcy@gmail.com (M. Nath).

http://dx.doi.org/10.1016/j.ica.2014.02.034
0020-1693/Ó 2014 Elsevier B.V. All rights reserved.

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
2 M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx

and several research groups are actively engaged in the field (±)-mandelic acid (HMal) and gallic acid (HGal) (Fig. 1), and inves-
[9–23]. Many organotin compounds have been established as tigate their biological activities, viz. in vitro anti-tumor activity
potential cancer chemotherapeutic agents because of their apopto- (against MCF-7, HEK-293, PC-3, HCT-15 and HepG-2), in vivo
tic inducing character [22–26], and they occupy an important place anti-inflammatory activity and toxicity studies. Furthermore, to
in cancer chemotherapy reports [11,12,17b,24]. Furthermore, the view insight the mode of action of the studied complexes various
National Cancer Institute has tested about 2000 tin-based assays, such as lactate dehydrogenase, glutathione reductase, glu-
compounds, the largest number ever tested among metal-based tathione peroxidase, total glutathione content, lipid peroxidase,
drug candidates [27], and recently it has been reported that DNA fragmentation, acridine orange and comet assay have also
four out of thirty interesting inorganic pharmaceuticals are tin been conducted and discussed in this manuscript.
compounds [28]. Despite this, the exact mechanism of mode of
action of organotin compounds remains unestablished.
Organotin(IV) carboxylates are among the most widely studied 2. Material and methods
class of compounds owing to their rich structural chemistry rang-
ing from monomeric, dimeric, tetrameric, oligomeric to polymeric 2.1. Materials
motifs [13,22,23,29–31]. The structures can be easily controlled by
adjusting the carboxylato ligands, tin–R groups and metal-to- The specifications and makes of triorganotin(IV) compounds,
ligand molar ratio [29,32,33]. In addition to their structural diver- and methods to obtain various dried solvents are same as reported
sity, the organotin(IV) carboxylates also present very promising previously [22]. Gallic acid, b-D-glucuronic acid and (±)-mandelic
antitumor activities [11,12,22,23,27,34–42]. Among these, several acid were procured from Alfa Aesar and used as received. Further,
di-n-butyltin(IV) carboxylates have been found to be the most a large number of chemicals used for biological activities are also
active, and in some cases they exhibit much higher activity than same as reported in our previous manuscript [22]. Five cancer cell
clinically used reference compounds such as cis-platin, doxorubi- lines of human origin, viz. MCF-7 mammary cancer, HEK-293 kid-
cine and methotrexate [11,12,27,37]. ney cancer, PC-3 prostate cancer, HCT-15 colon cancer and
The systemic toxicity of chemotherapeutic drugs, their lack of HepG-2 liver cancer were purchased from National Center for Cell
tumor localization and an even distribution throughout the body Science (NCCS) Pune, India. For anti-inflammatory activity and
including tumor tissues are the most significant challenges facing acute toxicity Swiss albino mice were procured from All India
effective cancer chemotherapy. Therefore, a rational drug design Institute of Medical Science (AIIMS) Delhi, India.
is essentially required to optimize specific targeting features and
to control toxicity (side effects), by controlling thermodynamic
2.2. Physical measurements
and kinetic processes of metal complexes viz., choice of metal ion
and its oxidation state [43] or tailoring of ligand scaffold. It is also
The melting points, microanalyses (C, H and N) and tin content
well known that the cytotoxicity is related to some extent to lipo-
were determined as described previously [22]. Molar conductance
philicity of the drugs i.e., the most lipophilic compounds are the
measurements, infrared and far-infrared, nuclear magnetic reso-
most cytotoxic [44]. Further, the ligands having biologically active
nance (1H, 13C and 119Sn) spectra were recorded on the same
pharmacophore with biocompatible properties are tethered to
instruments as reported previously [22]. ESI-MS spectra were
others having multifunctional N and O donors to obtain modulated
recorded on Bruker MicroTOF-Q II mass spectrometer at a flow rate
ligand scaffold which can mute the potential toxicity of metallo-
of 180 lL/h on positive mode in water–acetonitrile mixture.
pharmaceuticals drugs [8].
Optical density (OD) in MTT assay, gel electrophoresis, enzymes
Anti-tumor activity of several organotin compounds has been
assays, in vivo anti-inflammatory activity and toxicity were done
reported by various workers, but their solubility has always
according to the reported procedures with slight modifications as
remained a problem. This can be solved by choosing ligands having
described in our previous manuscript [22].
polar groups such as hydroxy groups, or other oxygen or nitrogen
containing groups or substituted fluorine which may enhance their 0,34024 g
solubility. Glucuronic acid (HGlu), mandelic acid (HMal) and gallic 2.3. Synthesis
acid (HGal) are such hydroxycarboxylic acids which are highly sol-
uble in water, methanol, ethanol and other organic solvents and 2.3.1. Synthesis of sodium salt of hydroxycarboxylic acids
possess various medicinal properties. Glucuronic acid (uronic acid) Hydroxycarboxylic acid (HGlu/HMal/HGal, 2.0 mmol) was
exhibits anti-oxidant activity [45]. Mandelic acid (a-hydroxyl car- dissolved in 10 mL of specially dried ethanol under dry nitrogen
boxylic acid) has been used for the treatment of skin problems, and and added to sodium ethoxide, prepared by reacting sodium
also possess anti-bacterial activity [46]. Gallic acid is a naturally (0.058 g, 2.5 mmol) with dry ethanol (25 mL). The resulting mix-
occurring plant phenol obtained by the hydrolysis of tannins and ture was refluxed giving a clear solution of sodium salt of the acid
is known to display some pharmacological activities such as anti- within half an hour. Refluxing was continued for another 1–2 h
cancer, fungicidal/fungi-static, antiviral, anti-inflammatory and with constant stirring. The solution was concentrated and solid
anti-oxidant properties [47]. was dried in vacuum.
Interaction of glucuronic acid with bis(tributyltin) oxide
(studied via NMR and mass spectroscopy) in solution [48], the
solution studies of tris(1-butyl)stannyl-D-glucuronate through 2.3.1.1. Na(Glu). IR (cm1): mas(OCO) 1616s, ms(OCO) 1429m, Dm
NMR spectral analysis [49], di-n-butyltin(IV) and diethyltin(IV) 187, mas(COC) 1245m, ms(COC) 1110m, m(OH-1) 3533s, m(OH-2)
complexes of methoxy analog of gallic acid [50], reaction of ethyl- 3400s, m(OH-3) 3488s, m(OH-4) 3509s. 1H NMR (500.13 MHz,
trichlorostannane with N,N-dimethylamide of mandelic acid [51] Bz-d6, ppm): d 4.63 (d, 1H, H-1, J1,2 = 8.0 Hz), 3.25 (dd, 1H, H-2,
and synthesis of trimethyltin derivative of mandelic acid [52] are J1,2 = 8.0 Hz, J2,3 = 9.1 Hz), 3.50 (dd, 1H, H-3, J2,3 = 9.1 Hz,
few reports available in literature. Moreover, the functional groups J3,4 = 9.2 Hz), 3.96 (dd, 1H, H-4, J3,4 = 9.2 Hz, J4,5 = 9.6 Hz), 3.93 (d,
attached to carboxylic acid may have pronounced influence 1H, H-5, J4,5 = 9.6 Hz), 4.81 (s, 1H, OH-1), 3.55 (s, 1H, OH-2), 3.72
(increase/decrease) on the biological properties of organotin(IV) (s, 1H, OH-3), 3.86 (s, 1H, OH-4). 13C NMR (125.75 MHz, Bz-d6,
carobxylates [22,34–37]. Therefore, it becomes indispensable to ppm): d 95.86 (C-1), 74.53 (C-2), 72.43 (C-3), 72.74 (C-4), 76.48
synthesize organotin(IV) derivatives of b-D-glucuronic acid (HGlu), (C-5), 175.31 (C-6).

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx 3

Fig. 1. Molecular structures of hydroxycarboxylic acids.

2.3.1.2. HGlu. IR (cm1): mas(OCO) 1596s, ms(OCO) 1423s, Dm 173, (s, 2H, H-3, H-7), 3.71 (s, 1H, OH-4), 3.82 (s, 1H, OH-5), 3.76 (s,
mas(COC) 1249m, ms(COC) 1115m, m(OH-1) 3530s, m(OH-2) 3405s, 1H, OH-6). 13C NMR (125.75 MHz, CD3OD, ppm): d 167.96 (C-1),
m(OH-3) 3486s, m(OH-4) 3511s. 1H NMR (500.13 MHz, Bz-d6, 128.48 (C-2), 129.84 (C-3, C-7), 137.60 (C-4, C-6), 140.05 (C-5).
ppm): d 12.27 (s, 1H, (COO)–H), 4.71 (d, 1H, H-1, J1,2 = 9.3 Hz),
3.35 (dd, 1H, H-2, J1,2 = 9.3 Hz, J2,3 = 9.0 Hz), 3.39 (dd, 1H, H-3, 2.3.2. Synthesis of triorganotin(IV) derivatives of hydroxycarboxylic
J2,3 = 9.0 Hz, J3,4 = 9.1 Hz), 3.85 (dd, 1H, H-4, J3,4 = 9.1 Hz, acids
J4,5 = 9.5 Hz), 3.76 (d, 1H, H-5, J4,5 = 9.5 Hz), 4.77 (s, 1H, OH-1), A hot ethanolic solution (20 mL) of trimethyltin(IV) chloride
3.49 (s, 1H, OH-2), 3.62 (s, 1H, OH-3), 3.90 (s, 1H, OH-4). 13C (0.398 g, 2.0 mmol) or tri-n-butyltin(IV) chloride (0.651 g,
NMR (125.75 MHz, Bz-d6, ppm): d 95.61 (C-1), 74.49 (C-2), 72.32 2.0 mmol) or triphenyltin(IV) chloride (0.771 g, 2.0 mmol) was
(C-3), 72.68 (C-4), 76.39 (C-5), 173.22 (C-6). added to the dry and hot ethanolic solution (10 mL) of the
preformed sodium salt of the acid as prepared in Section 2.3.1.
2.3.1.3. Na(Mal). IR (cm1): mas(OCO) 1597w, ms(OCO) 1420s, Dm The resulting solution was refluxed with constant stirring for an-
177, m(OH-2) 3610s. 1H NMR (500.13 MHz, CD3OD, ppm): d 5.11 other 5–6 h under dry nitrogen atmosphere. The resulting solution
(s, 1H, H-2), 7.73 (m, 1H, H-4), 7.31 (m, 2H, H-5 + H-7), 7.21 (m, was then centrifuged and filtered in order to remove the sodium
1H, H-6), 7.52 (m, 1H, H-8), 3.30 (s, 1H, OH-2). 13C NMR chloride formed during the reaction. The excess of solvent was re-
(125.75 MHz, CD3OD, ppm): d 175.33 (C-1), 75.22 (C-2), 136.19 moved under reduced pressure and the products thus obtained
(C-3), 127.21 (C-4), 127.41 (C-5), 126.21 (C-6), 127.33 (C-7), were washed with either ethanol–hexane or ethanol–petroleum
126.06 (C-8). ether (b.p. 40–60 °C) mixture (1:3 v/v).

2.3.1.4. HMal. IR (cm1): mas(OCO) 1580m, ms(OCO) 1417m, Dm 163, 2.3.2.1. Ph3Sn(Glu). White solid. M.P.: 98 °C. Yield: 60%. Anal. Calc.
m(OH-2) 3621m. 1H NMR (500.13 MHz, CD3OD, ppm): d 11.81 (s, for C24H24O7Sn: Sn, 22.92; H, 4.42; C, 53.07. Found: Sn, 22.99; H,
1H, (OCO)–H), 5.01 (s, 1H, H-2), 7.70 (m, 1H, H-4), 7.23 (m, 2H, 4.40; C, 53.01%. Mw: 543. IR (cm1): mas(OCO) 1626s, ms(OCO)
H-5 + H-7), 7.19 (m, 1H, H-6), 7.49 (m, 1H, H-8), 3.50 (s, 1H, OH- 1422s, Dm 204, mas(Sn–C) 277s, ms(Sn–C) 240s, mas(COC) 1150m, ms
2). 13C NMR (125.75 MHz, CD3OD, ppm): d 173.33 (C-1), 75.12 (COC) 1005m, m(Sn–O) 546w, m(OH-1) 3480br, m(OH-2) 3379w,
(C-2), 136.12 (C-3), 127.13 (C-4), 127.36 (C-5), 126.16 (C-6), m(OH-3) 3360br, m(OH-4) 3410m. 1H NMR (500.13 MHz, CD3OD,
127.29 (C-7), 125.96 (C-8). ppm): d 4.82 (d, 1H, H-1, J1,2 = 8.2 Hz), 3.37 (m, 1H, H-2), 3.40 (m,
1H, H-3); 3.97 (m, 1H, H-4), 3.86 (d, 1H, H-5, J4,5 = 9.3 Hz), 4.77
2.3.1.5. Na(Gal). IR (cm1): mas(OCO) 1629s, ms(OCO) 1403s, Dm 226, (s, 1H, OH-1), 3.94 (s, 1H, OH-2)a, 3.95 (s, 1H, OH-3), 3.99 (s, 1H,
(OH-4) 3533br, m(OH-5) 3440br, m(OH-6) 3459br. 1H NMR OH-4), 7.72 (d, 6H, H-b, J = 3.2 Hz), 7.51 (m, 6H, H-c), 7.32 (t, 3H,
(500.13 MHz, CD3OD, ppm): d 7.30, 7.89 (s, 2H, H-3, H-7), 3.81 (s, H-d, J = 6.5 Hz). 13C NMR (125.75 MHz, CD3OD, ppm): d 97.24 (C-
1H, OH-4), 3.71 (s, 1H, OH-5), 3.90 (s, 1H, OH-6). 13C NMR 1), 74.42 (C-2), 72.65 (C-3); 73.93b (C-4), 77.51 (C-5), 178.00 (C-
(125.75 MHz, CD3OD, ppm): d 169.68 (C-1), 127.98 (C-2), 130.01 6), 129.93 (C-a,1J(13C–119Sn) = 466.21 Hz, h = 117.65°), 138.12 (C-
(C-3, C-7), 137.89 (C-4, C-6), 140.13 (C-5). b, 2J(13C–119Sn) = 44.33 Hz), 129.52 (C-c, 3J(13C–119Sn) = 68.00 Hz),
130.24 (C-d). 119Sn NMR (186.50 MHz, CD3OD, ppm): d 141.44.
1
2.3.1.6. HGal. IR (cm1): mas(OCO) 1607m, ms(OCO) 1425 s, Dm 182, H NMR (500.13 MHz, CDCl3, ppm): d 4.82 (d, 1H, H-1,
(OH-4) 3540m, m(OH-5) 3509 w, m(OH-6) 3499m. 1H NMR J1,2 = 8.1 Hz), 3.32 (m, 1H, H-2), 3.35 (m, 1H, H-3), 3.86 (m, 1H,
(500.13 MHz, CD3OD, ppm): d 12.23 (s, 1H, (OCO)–H), 7.28, 7.85 H-4), 3.79 (d, 1H, H-5, J4,5 = 9.2 Hz), 3.99 (s, 1H, OH-1)a, 3.88

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
4 M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx

(s, 1H, OH-2), 3.93 (s, 1H, OH-3), 3.97 (s, 1H, OH-4), 7.73 (d, 6H, H- (s, 1H, H-2), 7.90 (m, 1H, H-4), 7.00 (m, 2H, H-5 + H-7), 6.98 (m,
b, J = 3.1 Hz), 7.50 (m, 6H, H-c), 7.33 (t, 3H, H-d, J = 6.5 Hz). 13C NMR 1H, H-6), 7.17 (m, 1H, H-8), 3.95 (s, 1H, OH-2), 7.60 (d, 6H, H-b,
(125.75 MHz, CDCl3, ppm): d 97.10 (C-1), 74.34 (C-2), 72.59 (C-3), 73.81b J = 3.1 Hz), 7.42 (m, 9H, H-c + H-d). 13C NMR (125.75 MHz, CD3OD,
(C-4), 77.32 (C-5), 176.45 (C-6), 129.80 (C-a, 1J(13C–119Sn) = 350.33 Hz, ppm): d 177.41 (C-1), 78.10 (C-2), 135.89 (C-3), 129.10 (C-4),
h = 107.46°), 138.00 (C-b, 2J(13C–119Sn) = 44.01 Hz), 129.13 (C-c, 3J 127.62 (C-5), 126.25 (C-6), 127.46 (C-7), 127.12 (C-8), 139.60 (C-
(13C–119Sn) = 67.71 Hz), 129.96 (C-d). 119SnNMR (186.50 MHz, CDCl3, a), 137.33 (C-b), 130.14 (C-c), 130.31 (C-d). 119Sn NMR
ppm): d –40.91. ESI-MS: 544 [M+H]+, 566 [M+Na]+ (100%), 350 (186.50 MHz, CD3OD, ppm): d 182.51. ESI-MS: 502 [M+H]+, 524
(ML)+, 274 [HSnPh2]+, 198 [H2SnPh]+. [M+Na]+ (100%), 350 (ML)+, 851 [M+SnPh3]+, 274 [HSnPh2]+,
198 [H2SnPh]+.
2.3.2.2. n-Bu3Sn(Glu). Creamish semi-solid. Yield: 62%. Anal. Calc.
for C18H36O7Sn: Sn, 24.64; H, 7.46; C, 44.75. Found: Sn, 24.68; H, 2.3.2.5. n-Bu3Sn(Mal). White solid. M. P.: 170°C. Yield: 69%. Anal.
7.50; C: 44.79%. Mw: 483. IR (cm1): mas(OCO) 1625m, ms(OCO) Calc. for C20H34O3Sn: Sn, 26.98; H, 7.71; C, 54.42. Found: Sn,
1415s, Dm 210, mas(Sn–C) 557sh, ms(Sn–C) 510s, mas(COC) 1161m, 26.93; H, 7.76; C, 54.49%; Mw: 441. IR (cm1): mas(OCO) 1609s,
ms(COC) 1015m, m(Sn–O) 560w, m(OH-1) 3494br, m(OH-2) 3373br, ms(OCO) 1396s, Dm 213, mas(Sn–C) 546s, ms(Sn–C) 515 m, m(Sn–O)
m(OH-3) 3436 w, m(OH-4) 3419br. 1H NMR (500.13 MHz, CD3OD, 562w, m(OH-2) 3271br. 1H NMR (500.13 MHz, CD3OD, ppm): d
ppm): d 5.01 (d, 1H, H-1, J1,2 = 8.1 Hz), 3.40 (m, 1H, H-2); 3.43 5.90 (s, 1H, H-2), 7.71 (m, 1H, H-4), 7.30 (m, 2H, H-5 + H-7), 7.22
(m, 1H, H-3), 3.89 (m, 1H, H-4), 3.79 (d, 1H, H-5, J4,5 = 9.0 Hz), (mbr, 1H, H-6), 7.60 (m, 1H, H-8), 3.88 (s, 1H, OH-2), 1.35 (t, 6H,
4.82 (s, 1H, OH-1), 3.83 (s, 1H, OH-2), 3.92 (s, 1H, OH-3), 3.98 (s, H-a, J = 6.7 Hz), 1.61 (m, 6H, H-b), 1.11 (m, 6H, H-c), 0.83 (t, 9H,
1H, OH-4)a, 1.83 (t, 6H, H-a, J = 6.8 Hz); 1.30 (m, 6H, H-b); 1.21 H-d, J = 6.7 Hz). 13C NMR (125.75 MHz, CD3OD, ppm): d 178.00
(m, 6H, H-c), 0.90 (t, 9H, H-d, J = 6.8 Hz). 13C NMR (125.75 MHz, (C-1), 79.21 (C-2), 136.11 (C-3), 129.13 (C-4), 127.41
CD3OD, ppm): d 97.89 (C-1), 74.41 (C-2), 72.65 (C-3), 73.16b (C- (C-5), 126.46 (C-6), 127.86 (C-7), 126.37 (C-8), 19.71 (C-a, 1J
4), 77.44 (C-5), 176.61 (C-6), 20.26 (C-a, 1J(13C–119Sn) = 454.43 Hz, (13C–119Sn) = 471.04 Hz, h = 118.07°), 27.22 (C-b), 26.73 (C-c),
h = 116.62°), 27.62 (C-b), 27.02 (C-c, 3J(13C–119Sn) = 82.00 Hz), 13.84 (C-d). 119Sn NMR (186.50 MHz, CD3OD, ppm): d 168.51.
14.21 (C-d). 119Sn NMR (186.50 MHz, CD3OD, ppm): d 168.10. ESI-MS: 442 [M+H]+, 464 [M+Na]+, 290 (ML)+ (100%), 731
1
H NMR (500.13 MHz, CDCl3, ppm): d 4.93 (d, 1H, H-1, [M+SnBu3]+, 234 [HSnBu2]+, 178 [H2SnBu]+.
J1,2 = 8.0 Hz), 3.35 (m, 1H, H-2), 3.36 (m, 1H, H-3), 3.95 (m, 1H,
H-4), 3.85 (d, 1H, H-5, J4,5 = 9.3 Hz), 4.07 (s, 1H, OH-1), 3.78 (s, 2.3.2.6. Me3Sn(Mal). White solid. M.P.: (123)d 118–120 °C; Yield:
1H, OH-2), 3.92 (s, 1H, OH-3), 3.97 (s, 1H, OH-4), 1.83 (t, 6H, H-a, 65% Anal. Calc. for C11H16O3Sn: Sn, 37.77; H, 5.08; C, 41.90. Found:
J = 6.7 Hz), 1.31 (m, 6H, H-b), 1.22 (m, 6H, H-c), 0.90 (t, 9H, H-d, Sn, 37.70; H, 5.09; C, 41.93%. Mw: 315. IR (cm1): mas(OCO) 1628s,
J = 6.8 Hz). 13C NMR (125.75 MHz, CDCl3, ppm): d 97.06 (C-1), ms(OCO) 1417m, Dm 211, mas(Sn–C) 601s, ms(Sn–C) 566s, m(Sn–O)
74.32 (C-2), 72.38 (C-3), 72.89b (C-4), 77.10 (C-5), 176.40 (C-6), 557w, m(OH-2) 3265br. 1H NMR (500.13 MHz, CD3OD, ppm): d
22.20 (C-a, 1J(13C–119Sn) = 334.65 Hz, h = 106.11°), 27.33 (C-b), 6.62 (s, 1H, H-2), 7.77 (m, 1H, H-4), 7.32 (m, 2H, H-5 + H-7), 7.25
26.82 (C-c, 3J(13C–119Sn) = 81.11 Hz), 12.11 (C-d). 119Sn NMR (m, 1H, H-6), 7.64 (m, 1H, H-8), 3.94 (s, 1H, OH-2), 0.80 (s, 9H,
(186.50 MHz, CD3OD, ppm): d 45.12. ESI-MS: 484 [M+H]+, 506 H-a, 2J(1H–119Sn) = 64.10 Hz, h = 114.93). 13C NMR (125.75 MHz,
[M+Na]+ (100%), 290 (ML)+, 234 [HSnBu2]+, 178 [H2SnBu]+. CD3OD, ppm): d 178.50 (C-1), 76.11 (C-2), 135.61 (C-3), 130.74
(C-4), 128.10 (C-5), 126.12 (C-6), 127.42 (C-7), 129.00 (C-8),
2.3.2.3. Me3Sn(Glu). White solid. M.P.: 73 °C. Yield: 58%. Anal. Calc. 16.85 (C-a, 1J(13C–119Sn) = 476.12 Hz, h = 118.52°). 119Sn NMR
for C9H18O7Sn: Sn, 33.35; H, 5.04; C, 30.25. Found: Sn, 33.38; H, (186.50 MHz, CD3OD, ppm): d 149.15. ESI-MS: 316 [M+H]+, 338
5.08; C, 30.28%. Mw: 357. IR (cm1): mas(OCO) 1626s, s(OCO) [M+Na]+, 164 (ML)+ (100%), 479 [M+SnMe3]+, 150 [HSnMe2]+,
1413m, Dm 213, mas(Sn–C) 590s, ms(Sn–C) 575s, mas(COC) 1154m, 136 [H2SnMe]+.
ms(COC) 1019m, m(Sn–O) 550w, m(OH-1) 3484br, m(OH-2) 3370br,
m(OH-3) 3429 w, m(OH-4) 3421br. 1H NMR (500.13 MHz, CD3OD, 2.3.2.7. Ph3Sn(Gal). White solid. M.P.: 221–226 °C. Yield: 61%. Anal.
ppm): d 5.08 (d, 1H, H-1, J1,2 = 8.3 Hz), 3.37 (m, 1H, H-2), 3.39 (m, Calc. for C25H20O5Sn: Sn, 22.93; H, 3.85; C, 57.80. Found: Sn, 22.87;
1H, H-3), 3.92 (m, 1H, H-4), 3.75 (d, 1H, H-5, J4,5 = 9.2 Hz,), 4.81 H, 3.89; C, 57.79%. Mw: 519. IR (cm1): mas(OCO) 1642 m, ms(OCO)
(s, 1H, OH-1), 3.89 (s, 1H, OH-2), 3.90 (s, 1H, OH-3), 3.91 (s, 1H, 1413s, Dm 229, mas(Sn–C) 288s, ms(Sn–C) 225m, m(Sn–O) 556w,
OH-4)a, 0.40, 0.11 (s, 9H, H-a, 2J(1H–119Sn) = 66.12 Hz, m(OH-4) 3444br, m(OH-5) 3440br, m(OH-6) 3409br. 1H NMR
h = 116.51°). 13C NMR (125.75 MHz, CD3OD, ppm): d 97.83 (C-1), (500.13 MHz, CD3OD, ppm): d 7.75, 7.88 (s, 2H, H-3, H-7)c, 4.07
74.45 (C-2) 72.26 (C-3), 75.16b (C-4), 77.41 (C-5), 178.92 (C-6), (s, 1H, OH-4), 3.79 (s, 1H, OH-5), 4.66 (s, 1H, OH-6), 8.20 (d, 6H,
1 13
4.3 (C-a, J( C–119Sn) = 447.10 Hz, h = 115.97° 119Sn NMR H-b, J = 3.3 Hz), 7.74 (m, 6H, H-c), 7.46 (t, 3H, H-d, J = 7.1 Hz). 13C
(186.50 MHz, CD3OD, ppm): d 183.01. 1H NMR (500.13 MHz, NMR (125.75 MHz, CD3OD, ppm): d 172.43 (C-1), 123.72 (C-2),
CDCl3, ppm): d 4.94 (d, 1H, H-1, J1,2 = 8.1 Hz), 3.37 (m, 1H, H-2), 110.02 (C-3), 144.04 (C-4), 141.41 (C-5), 137.87 (C-6), 109.00 (C-
3.39 (m, 1H, H-3), 3.90 (m, 1H, H-4), 3.70 (d, 1H, H-5, 7), 130.15 (C-a, 1J(13C–119Sn) = 484.57 Hz, h = 119.21°), 137.54 (C-
J4,5 = 9.3 Hz), 4.98 (s, 1H, OH-1), 3.82 (s, 1H, OH-2), 3.92 (s, 1H, b), 129.66 (C-c), 130.34 (C-d). 119Sn NMR (186.50 MHz, CD3OD,
OH-3), 3.98 (s, 1H, OH-4), 0.42, 0.09 (s, 9H, H-a, 2J(1H–119- ppm): d 132.19. ESI-MS: 520 [M+H]+, 542 [M+Na]+, 350 (ML)+
Sn) = 43.82 Hz, h = 106.47°). 13C NMR (125.75 MHz, CDCl3, ppm): d (100%), 869 [M+SnPh3]+, 274 [HSnPh2]+, 198 [H2SnPh]+.
97.78 (C-1), 74.05 (C-2), 72.00 (C-3), 75.00b (C-4), 77.24 (C-5),
177.86 (C-6), 2.2 (C-a, 1J(13C–119Sn) = 329.07 Hz, h = 105.62°]. 2.3.2.8. n-Bu3Sn(Gal). Yellowish solid. M.P.: 95 °C. Yield: 51%. Anal.
119
Sn NMR (186.50 MHz, CDCl3, ppm): d 49.21. ESI-MS: 358 Calc. for C19H32O5Sn: Sn, 25.93; H, 6.97; C, 49.67. Found: Sn, 25.90;
[M+H]+, 380 [M+Na]+, 164 (ML)+ (100%), 150 [HSnMe2]+, 136 H, 6.92; C, 49.63%. Mw: 459. IR (cm1): mas(OCO) 1637s, ms(OCO)
[H2SnMe]+. 1410s, Dm 227, mas(Sn–C) 550s, ms(Sn–C) 521s, m(Sn–O) 565w,
m(OH-4) 3446br, m(OH-5) 3437br, m(OH-6) 3412br. 1H NMR
2.3.2.4. Ph3Sn(Mal). White solid. M.P.: 190 °C. Yield: 61%; Anal. Calc. (500.13 MHz, CD3OD, ppm): d 7.70, 7.86 (s, 2H, H-3, H-7)c, 4.18
for C26H22O3Sn: Sn, 23.75; H, 4.39; C, 62.28. Found: Sn, 23.70; H, (s, 1H, OH-4), 3.77 (s, 1H, OH-5), 4.52 (s, 1H, OH-6), 1.05 (t, 6H,
4.33; C, 62.20%; Mw: 501. IR (cm1): mas(OCO) 1650w, ms(OCO) H-a, 2J(1H–119Sn) = 56.17 Hz, h = 110.05°), 1.56 (m, 6H, H-b), 1.21
1427s, Dm 223, mas(Sn–C) 291s, ms(Sn–C) 230s, m(Sn–O) 555w, (m, 6H, H-c), 0.87 (t, 9H, H-d, J = 6.7 Hz). 13C NMR (125.75 MHz,
m(OH-2) 3268br. 1H NMR (500.13 MHz, CD3OD, ppm): d 4.81 CD3OD, ppm): d 173.51 (C-1), 124.02 (C-2), 109.72 (C-3), 143.30

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx 5

(C-4), 141.63 (C-5), 137.47 (C-6), 109.06 (C-7), 21.05 (C-a), 29.16 10 mM Tris, 1 mM of EDTA (pH 8.5), and 50 lg/mL of RNAase for
(C-b), 27.35 (C-c:), 13.17 (C-d). 119Sn NMR (186.50 MHz, CD3OD, 1 h at 37 °C.
ppm): d 161.43. ESI-MS: 460 [M+H]+, 482 [M+Na]+, 290 (M-L)+ For agarose gel electrophoresis, 0.4 g of agarose was added to
(100%), 749 [M+SnBu3]+, 234 [HSnBu2]+, 178 [H2SnBu]+. 50 mL of 1x TAE buffer (containing tris base, acetic acid and EDTA)
and the mixture was heated in a microwave oven till the agarose
2.3.2.9. Me3Sn(Gal). White solid. M.P.: 230–235 °C. Yield: 68%. Anal. was completely dissolved. When the solution becomes lukewarm,
Calc. for C10H14O5Sn: Sn, 35.74; H, 4.20; C, 36.04. Found: Sn, 35.77; 2 lL of ethidium bromide (10 mg/mL stock) was added. This solu-
H, 4.26; C, 36.09%. Mw: 333. IR (cm1): mas(OCO) 1643s, ms(OCO) tion was then poured into the gel casting tray fitted with appropri-
1421m, Dm 222, mas(Sn–C) 587m, ms(Sn–C) 568 s, m(Sn–O) 551w, ate comb and allowed to solidify. This gel was used for the
m(OH-4) 3441br, m(OH-5) 3432br, m(OH-6) 3415br. 1H NMR submerged electrophoresis for separating the DNA samples using
(500.13 MHz, CD3OD, ppm): d 7.85, 7.92 (s, 2H, H-3, H-7)c, 4.17 suitably 1 or 0.5 TAE as the running buffer. DNA samples were
(s, 1H, OH-4), 3.75 (s, 1H, OH-5), 4.73 (s, 1H, OH-6), 0.04, 0.08 (s, mixed with gel loading dye and loaded into the wells along with
9H, H-a). 13C NMR (125.75 MHz, CD3OD, ppm): d 174.10 (C-1), the controls. Gel was run at 80 V for 1–2 h till the dye front reached
123.91 (C-2), 110.18 (C-3), 144.52 (C-4), 142.31 (C-5), 137.28 the last one-third of the gel and was observed in a UV illuminator.
(C-6), 109.36 (C-7), 4.12 (C-a, 1J(13C–119Sn) = 447.50 Hz,
h = 116.01°]. 119Sn NMR (186.50 MHz, CD3OD, ppm): d 202.77.
ESI-MS: 334 [M+H]+, 356 [M+Na]+, 164 (ML)+ (100%), 497 2.4.2. Acridine orange assay
[M+SnMe3]+, 150 [HSnMe2]+, 136 [H2SnMe]+. Acridine orange staining is one of the acceptable assays for
Mw: molecular weight; IR: s, strong; m, medium; w, weak; sh, evaluating the apoptosis by analysing the plasma-membrane
shoulder; br, broad; Dm = mas(OCO)  ms(OCO); permeability, nuclear morphology as well as the chromatin con-
NMR: hydrogen and carbon number according to Fig. 1; s, singlet; densation [55,56]. In the assay, the cancer cells were stained with
d, doublet; t, triplet; q, quartet; m, multiplet; br, broad; angle h acridine orange (AO)/ethidium bromide (EB) dye mixture (100 lg/
calculated from Eq.: \C–Sn–C = 0.0161|2J|2  1.32|2J| + 133.4 [32] mL of AO and 100 lg/mL of EB) and assay was performed according
and |(1J(13C119Sn)| = 11.4h  875; avery weak intensity signal; bbroad to the method described by Takahashi et al. [56]. Briefly, 0.5  106
signal; c the signals of the two hydrogens may interexchange; cancer (MCF-7) cells were seeded in a 6-well plate and incubated
d
According to Ref. [52]; with the respective IC50 value of n-Bu3Sn(Gal) and cis-platin
(positive control) for 24 h and then washed with PBS. After
washing 500 lL of AO/EB dye mixture dissolved in PBS was added
to the plate and the cells were observed under fluorescent
microscope (Zeiss, Axiovert 25, Germany).

2.4. Biological studies 2.4.3. Comet assay


Comet assay is a very simple technique yet highly sensitive to
The synthesized triorganotin(IV) hydroxycarboxylates have detect DNA damage in individual cells [57]. Cytotoxicity effect of
been screened for their in vitro anti-cancer (MTT assay) and n-Bu3Sn(Gal) (test compound) which leads to DNA damage was
in vivo anti-inflammatory activity, and have also been investigated also validated by comet assay (single cell gel electrophoresis).
for acute toxicity according to the reported procedures which are The assay was performed according to the method reported earlier
described in our previous manuscript [22]. Further, various en- [58] with slight modifications. In brief, 5  105 cancer (MCF-7)
zyme assays, viz. lipid peroxidase, glutathione peroxidase, glutathi- cells were treated with test compound and incubated for 24 h at
one reductase, total glutathione and lactate dehyrogenase assays 37 °C and then washed thoroughly with PBS. Fresh media was
against MCF-7 cell lines by n-Bu3Sn(Gal) were carried out to deter- added after proper washing and then the cells were harvested after
mine the effect of the synthesized complexes on cancerous cell 24 h. Slides were prepared with 1% normal melting agarose, where,
lines, compared to control group of cells (non-treated). The details 0.3  106 cells were mixed with 50 lL of 1% low melting agarose at
of methodologies employed are the same as reported previously 37 °C in PBS and sandwiched between the previously prepared
[22]. DNA cleavage, acridine orange and comet assays against slide with 1% normal melting agarose. The prepared slides were
MCF-7 by n-Bu3Sn(Gal) have also been carried out according to stored at 4 °C for 10 min before incubating them in lysis buffer con-
the procedures as described below. sisting 2.5 M of NaCl, 100 mM of EDTA (pH 10), 10 mM of Tris-base,
1% sodium lauryl sarcosinate and 1% triton X-100 for 2 h at 4 °C.
2.4.1. DNA fragmentation assay Electrophoresis was carried out for 5 min at 25 V in 300 mM of
Natural DNA fragmentation generally occurs during pro- NaOH and 1 mM of EDTA. After that the slides were neutralized
grammed cell death and is a landmark for apoptosis. The DNA with tris base at pH 7.5. The nuclei stained with ethidium bromide
cleavage pattern occurring due to cytotoxic effect of n-Bu3Sn(Gal) were visualized under fluorescent microscope.
(test compound) was analyzed through agarose gel electrophoresis
[53,54]. 3  106 cancer (MCF-7) cells were exposed to the IC50 va-
lue of the test compound. Cells were centrifuged and then washed 2.4.4. Statistical analysis
with phosphate buffer saline (PBS), and the pellet was lysed with The results (for biological data) have been expressed as
400 lL hypotonic buffer solution (containing 10 mM of tris (pH mean ± standard error and unpaired two tailed t-test has been
7.5), 1 mM of EDTA and 0.2% triton X-100) for 15 min at room tem- performed to measure the significance in the results obtained.
perature, and then centrifuged at 13 000 rpm for 15 min. 350 lL of Student’s t-test is a statistical method of testing hypotheses about
the supernatant was again lysed in 106 lL of second lysis buffer the mean of a small sample when the population standard devia-
(150 mM NaCl, 10 mM Tris–HCl (pH 8.0), 40 mM EDTA, 1% SDS tion is not known.
and 0.2 mg/mL of proteinase K, at final concentration) for 4 h at The unpaired two tailed t-test performed has been used to
37 °C. The DNA was extracted with phenol/chloroform/isoamyl analyse the significance level at p < 0.5 for anti-inflammatory activ-
alcohol (25:25:1 v/v/v), and the pellet thus obtained was washed ity and enzyme activity data. The statistically significant p value
with ethanol and resuspended for RNAase digestion in 15 lL of has been calculated through GraphPad Prism 5 version.

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
6 M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx

3. Results and discussion found to be comparable with that of sodium salt of the gallic acid,
which indicates that the carboxylate group is bidentate and
3.1. Synthetic aspects bridging [22,59,60]. The disappearance of a broad band (2833–
2957 cm1) in the spectra of the triorganotin(IV) hydroxycarboxy-
The reaction of triorganotin chloride with sodium salt of lates (which was present in the free acids as a weak intensity
hydroxycarboxylic acid (formed according to Eq. (1)) in a 1:1 M band), suggests the deprotonation of the free (COOH) group upon
ratio led to the formation of complexes according to Eq. (2). complexation. Furthermore, appearance of a m(Sn–O) at
556 ± 10 cm1 supports the bonding of carboxylate oxygen with
HL þ NaOEt !NaL þ EtOH ð1Þ
the tin atom. The ms(Sn–C) and mas(Sn–C) have been observed at
their corresponding frequencies and are in accordance with the lit-
HL ¼ HGlu; HMal and HGal
erature values [21–23]. The m(O–H) of complexes broadens and
shifts towards lower frequency, indicating the involvement of hy-
1:1
droxy (OH) group in intra-/intermolecular hydrogen bonding.
It has been observed from the IR spectra of R3Sn(Glu) that the
R3 SnCl þ NaL!R3 SnðLÞ þ NaCl ð2Þ
mas(COC) and ms(COC) shift towards lower frequencies in compari-
son to the free acids. On this basis, it can be inferred that the ring
R ¼ Me; n-Bu and Ph
oxygen may also be participating in weak interaction with organo-
The reaction in Eq. (1) required 1–2 h and Eq. (2) required 5–6 h tin(IV) moiety. The carboxylate groups in organotin(IV) carboxyl-
of refluxing. The resulting products were obtained in good yield ates generally adopt a bridge structure in the solid state, unless
(51–68%). From the analytical data it can be inferred that the the organic substituent at the tin atom is bulky or carboxylate
resulting triorganotin(IV) hydroxycarboxylates crystallized with group is branched at the a-carbon [29]. Mandelic acid (a-hydroxy-
1:1 stoichiometry. R3Sn(Glu) are soluble in water/DMSO mixture, carboxylic acid) is branched at a-carbon, therefore, due to its
methanol, ethanol, ether, DMSO, benzene and chloroform, whereas expected steric property; the bridging through carboxylate group
R3Sn(Mal) and R3Sn(Gal) are soluble in DMSO and water/DMSO can be excluded [29]. But there is a possibility of hydroxy (OH)
mixture, show low solubility in ethanol, methanol, and insoluble group participation in bridging or interaction with tin, which has
in non-polar solvents. The synthesized complexes are found been supported by the relative lowering of m(OH) of the complexes
to be stable towards air and moisture. Diorganotin(IV) derivatives (3265–3271 cm1) in comparison with free mandelic acid
of these hydroxycarboxylic acids have not been obtained (3621 cm1) [52].
in pure form under the conditions used for triorganotin(IV)
hydroxycarboxylates. 3.4. Multinuclear (1H, 13
C and 119
Sn) NMR spectral studies

3.2. Molar conductance measurements 3.4.1. 1H NMR spectra


The characteristic resonance peaks in the 1H NMR spectra of the
Very low molar conductance values of 103 M solutions (in mandelic and gallic acid and their triorganotin(IV) derivatives have
methanol) of the synthesized triorganotin(IV) hydroxycarboxylates been recorded in CD3OD. The 1H NMR spectra of b-D-glucuronic
are in the range 8–12 ohm1 cm2 mol1, which suggest their acid has been recorded in benzene-d6 (because of its poor solubility
non-electrolytic nature. in methanol) and the triorganotin(IV) derivatives of glucuronic
acid have been recorded in both CDCl3 and CD3OD. The absence
3.3. Infrared and far-infrared spectral studies of 1H NMR signal corresponding to acidic hydrogen (CO(OH)) in
the spectra of triorganotin(IV) hydroxycarboxylates, suggests the
The structures of hydroxycarboxylic acids (HGlu, HMal and coordination of triorganotin(IV) moiety with the carboxylate
HGal) raise several questions such as whether there is a coordina- group.
tion to tin through the carboxylic oxygen or the hydroxy oxygen, In the 1H NMR spectra of R3Sn(Glu) and R3Sn(Gal), the protons
what is the coordination number of tin atom due to these interac- of hydroxyl groups (OH) have been observed as a weak, broad sig-
tions, and the detailed geometry around the tin atom where the nal which either remained at nearly same resonance or deshielded,
possibility of isomerism exists [29,30]. Furthermore, due to poly- indicating intra-/intermolecular hydrogen bonding. In 1H NMR
dentate nature of these acids, it is essential to differentiate the spectra of R3Sn(Mal) a significant change in OH resonance has been
monomeric structures (formed through chelation/intra-molecular observed in comparison to that of free mandelic acid, which sug-
bonding) and polymeric structures (formed through bridging/ gests that the bonding of tin with oxygen atom of OH group may
intermolecular bonding), where the later may further adopt one/ also be possible along with carboxylate group. The resonances
two dimensional structure or cyclic oligomeric structure [29,30]. due to all the magnetically non-equivalent protons in the studied
The stretching frequencies of carboxylic group (m(OCO)) has complexes have been successfully identified, and the total
been utilized to confirm the mode and nature of coordination numbers of protons calculated from the integration curve have
through carboxylate oxygen (monodentate, bidentate or bridging) been found to be in good agreement with those calculated from
in the studied triorganotin(IV) hydroxycarboxylates. The IR the proposed molecular formula.
absorption spectra of the studied complexes indicate that the mas Further, the 2J(1H119Sn) coupling constants obtained from the
(OCO) shift towards higher frequencies, whereas the corresponding resolved satellites for Me3Sn(Mal) and n-Bu3Sn(Gal) are 64.10 Hz
ms(OCO) absorption frequencies either remain at the same value or and 56.17 Hz, respectively. These values suggest penta-coordi-
move to lower frequencies in comparison to free acids. The magni- nated geometry for the complexes. The \CSnC calculated using
tude of (mas–ms)OCO (Dm) has been found to be useful in the identi- Lockhart and Manders equation [61] are 115.39° and 110.05° for
fication of structural features [22,23,59,60]. For R3Sn(Glu) and Me3Sn(Mal) and n-Bu3Sn(Gal), respectively. These values further
R3Sn(Mal), Dm (200 cm1) has been observed to be greater than suggest that the complexes exhibit trigonal–bipyramidal geome-
that of the sodium salt of the corresponding acid, which indicates try, with more electronegative (oxygen) groups in axial position
that the carboxylate group is monodentate [21,59,60] but bridging and organic groups in equatorial plane. The 2J(1H119Sn) values for
or chelation through other groups of the ligands cannot be ex- Me3Sn(Glu) have been observed at 43.82 Hz in CDCl3, while at
cluded. The magnitude of Dm for R3Sn(Gal) (>200 cm1) has been 66.12 Hz in CD3OD. These values indicate the four-coordinated

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx 7

geometry for Me3Sn(Glu) in non-coordinating solvent and midal geometry about tin. The proposed structures for the studied
five-coordinated geometry in coordinating solvent. The \CSnC triorganotin(IV) hydroxycarboxylates have been presented in
calculated for Me3Sn(Glu) in CDCl3 (106.47°) and CD3OD Fig. 2.
(116.51°) further support pseudo-tetrahedral geometry in CDCl3
and distorted trigonal–bipyramidal geometry in CD3OD. 3.5. ESI-MS spectrometric studies

3.4.2. 13C NMR spectra The ESI-MS spectral data for triorganotin(IV) hydroxycarboxy-
From 13C NMR spectral data of hydroxycarboxylic acids and lates have been recorded in the range m/z 50–1000, using metha-
their triorganotin(IV) derivatives it has been inferred that the nol as solvent and further diluting with acetonitrile and water
signals attributable to –(OCO) show significant downfield shift mixture. In the ESI-MS spectra of triorganotin(IV) hydroxycarboxy-
upon complex formation, which suggests coordination of triorga- lates, the weak molecular ion peak ([M+H]+) appears, which has
notin(IV) moiety with carboxylate group. been used for determining the molecular weight (Mw) of the com-
R3Sn(Glu) shows an interesting coordination behavior. The plexes. Mw for Ph3Sn(Glu), n-Bu3Sn(Glu) and Me3Sn(Glu) have
carbon atom C4 exhibits downfield shift, while C2 remaines unaf- been observed at 543, 483 and 357, respectively; for Ph3Sn(Mal),
fected, and C1 and C3 show only slight change in the chemical n-Bu3Sn(Mal) and Me3Sn(Mal) at 501, 441 and 315, respectively;
shifts. A small downfield shift for the C5 has also been observed, and for Ph3Sn(Gal), n-Bu3Sn(Gal) and Me3Sn(Gal) at 519, 459,
indicating that weak interaction of ring oxygen with triorgano- and 333, respectively. The observed molecular weights (Mw) of tri-
tin(IV) moiety may be possible [49,50]. However, the shifts are organotin(IV) hydroxycarboxylates are in excellent agreement
very small and hence the interactions between ring oxygen and with the calculated values, which verify the proposed molecular
tin atom can be excluded. formula for the complexes. In the ESI-MS spectra of triorgano-
In R3Sn(Mal), the C2 shows a significant downfield shift, which tin(IV) hydroxycarboxylates, [SnR3]+ ion peaks have been observed,
supports the weak interaction between oxygen of hydroxyl group which are due to loss of one mole of ligand from one mole of com-
(OH group) and tin atom. All the other carbon atoms of the acid plex, involving the cleavage of weak Sn–O bond yielding two com-
either remained unaffected or very small change in chemical shifts plementary ions, where the cationic part of the complex is
have been observed for them. A significant change in the chemical measured in positive mode [63]. Triorganotin(IV) hydroxycarboxy-
shift of C4 and C6 in 13C NMR spectra of R3Sn(Gal) has been ob- lates often adopt polymeric structure with five-coordinated tin and
served, which suggests hydrogen bonding through OH groups at the peak corresponding to [M+SnR3]+ provides additional proof for
meta positions of the acid. penta-coordinated linear polymeric motif [64]. The [M+SnR3]+
The 13C NMR chemical shifts of methyl, n-butyl and phenyl peaks have also been observed for R3Sn(Mal) and R3Sn(Gal), which
carbons attached to tin are observed at comparable positions as re- suggest penta-coordinated polymeric nature of the complexes. The
ported previously. The heteronuclear coupling constant subsequent fragment ions [HSnR2]+ and [H2SnR]+ resulted from
1 13
J( C–119Sn) provides useful information about the coordinating [SnR3]+ have been reported earlier in the literature for other
environment and geometry of organotin(IV) complexes. The satel- triorganotin(IV) carboxylates [65], similar peaks have also been ob-
lites have been resolved for most of the complexes and the served in ESI-MS spectra of the synthesised triorganotin(IV)
observed coupling constants 1J(13C–119Sn) are found to be in the hydroxycarboxylates. The ESI-MS spectrometric data do not pro-
range 465.60 ± 20 Hz. These values suggest penta-coordinated vide any crucial information about the structure of the complexes,
organotin moiety for R3Sn(Mal) and R3Sn(Gal). Further, the calcu- except that R3Sn(Mal) and R3Sn(Gal) may be polymeric in nature,
lated values of \CSnC (116.01–119.21°) with the help of Lockhart however the data helped in confirming the Mw and empirical for-
and Manders equation [62] {Eq. 1J(13C–119Sn) = 11.4h  875}, sug- mula of the complexes.
gest distorted trigonal–bipyramidal geometries for the complexes.
The 1J(13C–119Sn) values for R3Sn(Glu) have been observed in 3.6. Biological studies
the range 329.07–350.33 Hz in CDCl3, while 454.43–466.21 Hz in
CD3OD, indicating four-coordinated geometry in non-coordinating All the three hydroxycarboxylic acids viz. glucuronic acid,
solvent and five-coordinated geometry in coordinating solvent for mandelic acid and gallic acid are biologically important molecules
the complexes. The \CSnC calculated for R3Sn(Glu) in CDCl3 and individually known for their therapeutic applications. Thus, it
(107.46–105.61°) and CD3OD (117.68–115.97°) further support becomes indispensable to evaluate the biological behavior of syn-
the pseudo-tetrahedral geometry in CDCl3 and distorted trigonal– thesized triorganotin(IV) hydroxycarboxylates especially for their
bipyramidal geometry in CD3OD. anti-cancer and anti-inflammatory activities.

3.4.3. 119Sn NMR spectra 3.6.1. Cytotoxicity assay (MTT assay)


The 119Sn NMR shifts are very sensitive to coordination number To study the cytotoxicity effect of synthesized complexes, all of
of tin and greatly shift upfield on bonding to Lewis base. The 119Sn the triorganotin(IV) hydroxycarboxylates along with standard
chemical shifts increase markedly with the increase in coordina- drugs, viz. cis-platin and 5-fluorouracil have been screened
tion number, ca. d 50 to +200 ppm for four-coordinated tin; d in vitro against five cell lines of human origin, viz. MCF-7 (mam-
100 to 200 ppm for five-coordinated tin; d 200 to 330 ppm mary cancer), HEK-293 (kidney cancer), PC-3 (prostate cancer),
for six-coordinated tin compounds [22]. HCT-15 (colon cancer) and HepG-2 (liver cancer) and their
The 119Sn chemical shifts for R3Sn(Glu) have been observed in observed IC50 values are presented in Table 1. n-Bu3Sn(Gal) has
the region 40.91 to 49.21 ppm in CDCl3, suggesting four-coordi- been observed to be the most active among the studied triorgano-
nated tin. However, these complexes exhibit five-coordinated tin tin(IV) hydroxycarboxylates against all cancer cell lines, and it
in CD3OD (141.44 to 183.01 ppm), suggesting that these exhibits even higher activity in comparison to cis-platin, whereas
complexes exhibit additional coordination through inter- or it shows lower potential in comparison to 5-fluorouracil against
intramolecular association/interaction or from complexation with all cell lines except HCT-15. Ph3Sn(Glu) exhibits lowest activity
coordinating solvent [50]. amongst the studied complexes and lower potential in comparison
The 119Sn NMR shifts for R3Sn(Mal) and R3Sn(Gal) are observed to cis-platin as well as 5-fluorouracil.
in the region 202.77 to 132.19 ppm. These chemical shifts It has been reported [66] that the compounds exhibiting IC50 in
clearly suggest that they exhibit five-coordinated trigonal–bipyra- the range 1.0–10.0 lg/mL are cytotoxic and 10.0–30.0 lg/mL are

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
8 M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx

Fig. 2. Probable structures of triorganotin(IV) hydroxycarboxylates: (a) triorganotin(IV) glucuronates in coordinating solvent (R3Sn(Glu)solvent), (b) triorganotin(IV)
mandelates (R3Sn(Mal)) and (c) triorganotin(IV) gallates (R3Sn(Gal)); where R = Ph, n-Bu and Me.

Table 1
In vitro cytotoxicity data (IC50 expressed as lg/mL ± SEM) of triorganotin(IV) hydroxycarboxylates.

Complex MCF-7 HEK-293 PC-3 HCT-15 HepG-2


Ph3Sn(Glu) 29.95 ± 0.6 29.93 ± 0.8 26.66 ± 1.1 27.96 ± 0.6 28.46 ± 0.8
n-Bu3Sn(Glu) 22.77 ± 2.2 21.22 ± 1.7 20.78 ± 0.8 23.99 ± 0.6 20.74 ± 0.4
Me3Sn(Glu) 23.26 ± 2.5 25.55 ± 1.5 21.50 ± 3.1 25.21 ± 1.1 25.53 ± 1.4
Ph3Sn(Mal) 13.36 ± 1.1 16.10 ± 0.8 17.60 ± 1.1 17.02 ± 1.1 23.83 ± 0.3
n-Bu3Sn(Mal) 10.18 ± 1.3 10.15 ± 1.4 11.38 ± 0.8 12.70 ± 1.1 20.99 ± 0.3
Me3Sn(Mal) 12.62 ± 0.9 11.36 ± 0.8 16.0 ± 0.1 15.76 ± 0.4 21.19 ± 1.2
Ph3Sn(Gal) 9.03 ± 0.9 5.99 ± 0.7 6.45 ± 0.7 12.99 ± 0.3 18.24 ± 1.7
n-Bu3Sn(Gal) 2.04 ± 0.1 5.16 ± 0.4 5.30 ± 0.2 3.16 ± 0.5 3.98 ± 0.2
Me3Sn(Gal) 6.81 ± 0.9 6.28 ± 0.5 5.85 ± 0.8 11.70 ± 1.1 15.84 ± 0.3
Cis-platin 8.97 ± 0.10 6.72 ± 0.13 5.99 ± 0.05 3.71 ± 0.05 5.97 ± 0.10
5-Fluorouracil <0.65 1.61 ± 0.05 1.41 ± 0.05 7.17 ± 0.13 <0.65

moderately cytotoxic in nature. Triorganotin(IV) hydroxycarboxy- xybenzo-18-crown-6 and -15-crown-5 are less active than the
lates exhibit activity in the range 4–30 lg/mL. Therefore, it can corresponding triphenyltin derivatives but they are more active
be concluded that the complexes are cytotoxic to moderately cyto- than di-n-butyltin analogues [12b]. Triphenyltin derivatives of
toxic in nature. R3Sn(Gal) (gallic acid derivatives) are cytotoxic substituted-phenylacetic acid/-furoic acid/1,4-benzodioxane-6-
against MCF-7, HEK-293 and PC-3 and moderately cytotoxic carboxylic acid are more active than diphenyltin analogues [39].
against HCT-15 and HepG-2. R3Sn(Mal) and R3Sn(Glu) (mandelic However, in the present study tri-n-butyltin carboxylates are more
and glucuronic acid derivatives) exhibit moderate activity against active than the corresponding trimethyltin derivatives which are in
all cell lines. It has been observed that the activity increases as turn more active than the corresponding triphenyltin derivatives.
the alkyl carbon chain length increases, and further, the alkyl The highest activity of the tri-n-butyltin carboxylates may also
derivatives are more active than phenyl derivatives, similar results be related to the most lipophilic character of the n-butyl groups
have also been reported earlier [27]. as the lipophilicity of the more stable C-bound groups on tin is
The first systematic structure–activity relationships on several important in controlling their toxicity [27]. Further, the studied tri-
di- and tri-organotin(IV) carboxylates made by Gielen and his organotin derivatives of gallic acid are more active than those of
co-workers [12,27] reveals that di-n-butyltin(IV) carboxylates are mandelic acid which are in turn more active than those of glucu-
the most active compounds, and some of them exhibit much high- ronic acid.
er potency [11,12,27,37]. Further, tri-n-butyltin difluorobenzoates The structure–activity correlation for the studied triorgano-
are less active than the corresponding triphenyltin derivatives tin(IV) hydroxy-carboxylates reveals that the substituent attached
and also less active than the corresponding di-n-butyltin to OAC@O group has pronounced effect on the activity. Moreover,
derivatives [12a]. Similarly, tri-n-butyltin 3,6-dioxaheptanoates/ the activity of organotin compounds also depends on availability of
3,6,9-trioxadecanoates and tri-n-butyltin derivatives of 4-carbo- coordination positions around the tin atom, stability of the Sn–O

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx 9

bond towards hydrolytic cleavage (as well as Sn–C stability, phenyl


derivatives are prone to hydrolytic cleavage than alkyl ones), and
lipophilic and hydrophilic properties of the complexes [24,27,29].
Further, the conformation assumed by the ligand in the cytoplasm
may play an important role in activity of the complexes. Hence, the
transportation of R3Sn+ moiety or triorganotin(IV) hydroxycarb-
oxylate as a single entity across the cellular membrane may be
responsible for their activity.

3.6.2. Enzymatic assays


The observed data for enzymatic assays of n-Bu3Sn(Gal) against
MCF-7 are presented in Table 2. The graphical representation of the
data is expressed in Fig. 3–5. Necrosis and apoptosis are two ways
through which cell death can occur. Anti-tumor compounds gener-
ally arrest the cell growth through apoptosis. Apoptosis is a mode
of cell death that differs from necrosis. Apoptosis is PCD
Fig. 4. Total glutathione content in MCF-7 after the treatment with n-Bu3Sn(Gal)
(programmed cell death) while necrosis is PMCD (premature cell (mean ± SEM).
death).
Non-specific necrosis leads to cell death with in short interval of
time, mediated through membrane toxicity, which results in
sudden and marginal increase in lactate dehydrogenase (LDH)

Table 2
Enzymatic assays of n-Bu3Sn(Gal) against MCF-7.

Enzymes Activity
Untreated Treated Induction fold
a
Lipid peroxidation 4.58 ± 0.65 9.89 ± 0.11 2.16
Glutathione peroxidaseb 8.31 ± 0.03 9.81 ± 0.06 1.18
Total Glutathionec 6.31 ± 0.57 6.52 ± 1.25 1.03
Glutathione reductaseb 6.31 ± 0.027 4.95 ± 0.12 0.78
Lactate dehydrogenaseb 7.82 ± 0.03 8.05 ± 0.18 1.03
a
nmol/mg protein.
b
lmol/mg protein/min.
c
ng/mg protein; Induction fold: (mean of treated/mean of untreated); the
experiment was performed in triplicate and mean ± SEM of three independent
experiments are shown here.
Fig. 5. Lipid peroxidation in MCF-7 after the treatment with n-Bu3Sn(Gal)
(mean ± SEM). ⁄Represents the significant increase in lipid peroxidation with
respect to the control (p < 0.05), for MCF-7.

concentration. The result shows a very small increment in LDH


concentration, therefore, a minor role of necrosis in causing cell
death cannot be excluded. Substantial folds increase in enzymatic
activity suggests the generation of reactive oxygen species (ROS) in
the cells, leading to oxidative stress, which may be responsible for
cytotoxicty.
Lipid peroxidation is a common assay to measure oxidative
damage. n-Bu3Sn(Gal) treatment to MCF-7 cells results in high lipid
peroxidation which initiates the oxidative reactions, ensuring the
generation of reactive oxygen species (ROS) in the cells. The exces-
sive ROS generation may be responsible for cytotoxicity of the
complex. Various drugs are known in literature [67,68], which
disturb the intracellular mitochondrial membrane potential via
generation of reactive oxygen species (ROS).
Redox system of cells is regulated by certain enzymes such as
glutathione peroxidase and glutathione reductase [69]. Any change
in enzyme concentration of these enzymes may affect the
oxidative balance of cells and consequently subjecting the cells
to oxidative stress [70] and it is reported that the oxidative stress
plays an important role in apoptosis [71]. Induction of apoptosis
Fig. 3. Enzymatic activity: lactate dehydrogenase (LDH); glutathione peroxidase due to increase in glutathione peroxidase activity has also been
(GPx); glutathione reductase (GR) in MCF-7 after the treatment with n-Bu3Sn(Gal).
The results are expressed as mean ± SEM. ⁄⁄ and ## represent the significant
reported in literature [72]. Glutathione peroxidase (GPx), glutathi-
difference in glutathione reductase and glutathione peroxidase activity with respect one reductase (GR) activity and total glutathione content show
to control (p < 0.05), respectively. alteration in their activity for MCF-7 cells treated with n-Bu3-

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
10 M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx

Sn(Gal) with respect to control. Thus, n-Bu3Sn(Gal) may lead cell


death through induction of apoptosis.
Further, there are various mechanisms through which cell death
may be induced. One of them is cell membrane damage, resulting
in necrosis. Oxidative stress is the other possible way which may
affects cell survival and hence inhibiting cell growth. Alternatively,
n-Bu3Sn(Gal) might has induced peroxides generation, which are
metabolized by glutathione peroxidase (GPx), consequently form-
ing oxidized glutathione (GSSG) from reduced form of glutathione
(GSH). Decrease in GSH concentration and NADPH oxidation are
related to alteration of Ca2+ homeostasis, which is a crucial event
in loss of cell viability induced by peroxides [73]. Organotins also
influence [Ca2+] and mitochondrial activity, resulting in oxidative
stress in tumour cells leading to apoptosis [74]. Alteration in mito-
chondrial membrane potential may also be responsible for causing
apoptosis [75].
From the observed results of the enzyme assays it may be in-
ferred that both necrosis and apoptosis (apnosis) are responsible
for cell toxicity. Many anti-cancer drugs like cladribine, 5-fluoro-
uracil, doxorubicin, phyllanthus and cis-platin, have been reported
which lead to cell death through both apoptosis and necrosis [76].

3.6.3. DNA fragmentation assay


Natural DNA fragmentation generally occurs during pro-
grammed cell death and is a landmark for apoptosis [22]. DNA
fragmentation assay is among the most reliable method for distin-
guishing apoptosis from necrosis. DNA ladder/marker of 100 base
pair (lane 1), DNA extracted from untreated MCF-7 cells (lane 2),
DNA extracted from MCF-7 cells treated with n-Bu3Sn(Gal) (lane
3) and DNA extracted from MCF-7 cells treated with cis-platin as
reference drug (lane 4) were taken for DNA fragmentation assay
(as in Fig. 6). Gel electrophoresis experiment was conducted and
Fig. 6. DNA ladder formation through gel electrophoresis in MCF-7 cells; lane 1: recorded on Biorad-UV-transilluminator after the addition of
DNA marker; lane 2: control; lane 3: n-Bu3Sn(Gal); lane 4: cis-platin. ethidium bromide. The image was taken with the help of Gel
Doc-software. DNA–ladder formation (smearing) has been

Fig. 7. Acridine orange assay for (a) MCF-7 cells (control), (b) cis-platin treated MCF-7 cells, (c) n-Bu3Sn(Gal) treated MCF-7 cells. Unfilled arrows and filled arrows indicate
live cells and apoptotic cells, respectively. The experiment was performed in triplicate and a representative experiment is presented.

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx 11

observed for n-Bu3Sn(Gal) treated MCF-7 cells (Fig. 6), which sug- under fluorescent microscope. The Fig. 8 represents the single cell
gests that apoptotic pathway is mainly responsible for the cell gel (alkaline) electrophoresis showing DNA fragmentation in MCF-
death. 7 cells treated with n-Bu3Sn(Gal) in comparison to cis-platin and
control. The DNA breaks or fragmentation from individual cell
3.6.4. Acridine orange assay has been observed as comet tails (marked by arrows). However,
Morphological changes have been observed by acridine orange
fluorescent staining assay [55]. The Fig. 7 represents the induction
of apoptosis followed by membrane blebbing and chromatin
condensation by n-Bu3Sn(Gal) on MCF-7 cells. The cells were
incubated for 24 h with IC50 value of test compound and cis-platin.
Nuclear staining was visualized under fluorescent microscope
(Zeiss, Axiovert 25, Germany). n-Bu3Sn(Gal) induced cell death
has been considered to be apoptotic by simply observing the typi-
cal morphological change in the cells due to AO/EB staining. Live
cells and apoptotic cells can be easily distinguished by the percent-
age uptake of AO/EB but necrosis cannot be excluded completely.
In view of the fact that AO permeates all live cells, and hence ap-
peared green. EB is taken up only when the cells have lost their
cytoplasmic membrane integrity and hence appear red. It has been
observed (Fig. 7) that live cells have green nucleus (shown by un-
filled arrows), while apoptotic cells have orange nucleus (shown by
filled arrows).

3.6.5. Comet assay


Cytotoxicity leading to DNA damage has also been validated by
single cell gel electrophoresis. Comet assay or single cell gel elec-
trophoresis is simple yet sensitive technique which detects DNA
damage to the level of single cell [57]. The nuclei of MCF-7 cells Fig. 9. Percentage of DNA in comet tail in n-Bu3Sn(Gal) treated MCF-7 cells (A) in
(treated as well as control) were stained with EB and visualized comparison to cis-platin (B) and control (C).

Fig. 8. Comet assay for n-Bu3Sn(Gal) treated MCF-7 cells in comparison to cis-platin and control. The DNA breaks from individual cells were visualized as comet tails (marked
by arrows). The experiment was performed in triplicate and a representative experiment is presented.

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
12 M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx

Table 3
Anti-inflammatory activity and toxicity data of triorganotin(IV) hydroxycarboxylates.

Complex/standard drug Anti-inflammatory activity dose (50 mg/kg) Toxicity LD50


b (mg/kg)
Paw vol. before (A) Paw vol. after (B) Paw vol. difference p Value % Inhibition
(mean ± SEM) (mean ± SEM) (mean ± SEM)
Ph3Sn(Glu) 8.52 ± 0.28 9.92 ± 0.20 1.40 ± 0.11 0.0024 28.57 >200
n-BuSn(Glu) 8.30 ± 0.24 10.06 ± 0.16 1.76 ± 0.15 0.0005 10.20 >400
Me3Sn(Glu) 8.06 ± 0.10 9.86 ± 0.17 1.80 ± 0.17 0.0007 8.16 >200
Ph3Sn(Mal) 8.48 ± 0.25 9.98 ± 0.34 1.50 ± 0.16 0.0038 23.47 >600
n-Bu3Sn(Mal) 8.56 ± 0.24 10.30 ± 0.38 1.74 ± 0.18 0.0013 11.22 >600
Me3Sn(Mal) 8.26 ± 0.29 10.14 ± 0.28 1.88 ± 0.16 0.0003 4.08 >600
Ph3Sn(Gal) 8.24 ± 0.16 9.58 ± 0.14 1.34 ± 0.09 0.0015 31.6 3 >600
n-Bu3Sn(Gal) 8.40 ± 0.21 10.06 ± 0.61 1.66 ± 0.18 <0.0001 15.31 >800
Me3Sn(Gal) 8.20 ± 0.26 10.04 ± 0.32 1.84 ± 0.09 0.0004 6.12 >600
Ibuprofen 9.30 ± 0.23 10.64 ± 0.26 1.34 ± 0.13 – 36.19 >2000
Indomethacina 9.00 ± 0.35 9.82 ± 0.38 0.82 ± 0.09 – 60.95 >1000
Ph3SnCl 9.76 ± 0.11 11.68 ± 0.19 1.92 ± 0.08 – 8.57 >400
n-Bu3SnCl 9.46 ± 0.16 11.46 ± 0.31 2.00 ± 0.07 – 4.76 >400
Me3SnCl 9.70 ± 0.23 11.60 ± 0.18 1.90 ± 0.05 – 9.52 >400
a
Dose (5 mg/kg); paw volume before (A) and after (B) carrageenan injection.
b
calculated through unpaired t-test at significant level p < 0.05 with respect to Indomethacin.

vehicle treated cells (control) do not show any fragmentation. 4. Conclusions


Induction of DNA fragmentation (as shown in Fig. 8) has been
further validated by the comet assay. Triorganotin(IV) hydroxycarboxylates have been synthesized
Fig. 9 represents the % DNA in comet tail (58%) in MCF-7 cells through the sodium salt method. IR, 1H, 13C and 119Sn NMR spectral
treated with n-Bu3Sn(Gal). The results suggest that n-Bu3Sn(Gal) and ESI-MS studies of all the synthesized triorganotin(IV) hydroxy-
treated MCF-7 cells show a high level of DNA tailing in comparison carboxylates suggest a distorted trigonal–bipyramidal geometry.
to cis-platin (51%) treated cells and control (1.32%) cells. The Anti-cancer screening of these triorganotin(IV) hydroxylcarboxy-
results further support that apoptosis is the main cause for cell lates suggests that n-Bu3Sn(Gal) is the most active complex among
death. the studied complexes in this manuscript, which exhibits potent
% DNA = TA  TAI  100/[TA  TAI) + (HA  HAI)]; where anticancer activity against all the cell lines. Significant increase in
TAI = tail area intensity, HAI = head area intensity, TA = tail area glutathione peroxidase activity and lipid peroxidation assay sup-
and HA = head area [77]. port that cells undergo oxidative stress. Marginal increase in LDH
suggests that necrosis may also occur to a small extent. Further-
more, DNA ladder formation, AO and comet assays indicate that
3.6.6. Anti-inflammatory activity these complexes inhibit the cancer cell growth through apoptosis.
Cancer is related to inflammation to the some extent and there- Taken together, the results indicate that the complexes possess
fore, it was intriguing to see if the studied complexes also exhibit dual capability of cell death: necrosis (minor) and apoptosis
anti-inflammatory activity. The anti-inflammatory activity data (major). However, further experiments are needed to establish
(% inhibition) of synthesized triorganotin(IV) hydroxycarboxylates the fact more strongly. An anti-cancer agent is expected to possess
are presented in Table 3. The results obtained indicate the order of an anti-inflammatory activity. It has been observed that title com-
the anti-inflammatory activity of the studied triorganotin(IV) plexes show good anti-inflammatory activity. Among the studied
hydroxycarboxylates as: Ph3Sn(Gal) > Ph3Sn(Glu) > Ph3Sn(Mal) > complexes, Ph3Sn(Gal) exhibits the highest anti-inflammatory
n-Bu3Sn(Gal) > n-Bu3Sn(Mal) > n-Bu3Sn(Glu) > Me3Sn(Glu) > Me3 activity followed by Ph3Sn(Glu). In vitro cytotoxicity assays,
Sn(Gal) > Me3Sn(Mal). in vivo anti-inflammatory activity and toxicity results of the stud-
Ph3Sn(Gal) and Ph3Sn(Glu) have been found to exhibit potential ied complexes predict their potency as metal based anti-cancer
anti-inflammatory activity (31.63% and 28.57% inhibition, respec- drugs.
tively). Ph3Sn(IV)+ moiety, is membrane-active and highly lipo-
philic, can interact with lipid fraction and cytoplasmic Acknowledgements
membrane. The swift and frequent formation of Ph3Sn(IV)+ and
its transportation across the cellular membrane as a part of the We are thankful to technical staff of the Chemistry Department,
mechanism may be responsible for the higher efficacy [21–23]. I.I.T. Roorkee, for spectroscopic studies and elemental analysis.
Thanks are also due to The Head I.I.C. for providing NMR facility.
Ms. Monika Vats is thankful to MHRD, New Delhi, for financial
3.6.7. Toxicity Assistance.
The LD50 values of the studied triorganotin(IV) hydroxycarboxy-
lates are found to be in the range >200 to >800 mg/kg in albino References
mice (Table 3), which suggest that these complexes have moderate
toxicity with minimum adverse effects and wider safety margin. [1] K. Zdenek, K. Robert, B. Tomas, K. Jarmila, M. Pavel, K. Vladimir, Supramol.
This property is desirable from pharmaco-economic point of view. Chem. 24 (2012) 106.
[2] <http://www.drugsindex.htm>; <http://www.drugs.com>.
[3] B. Rosenberg, L. Vancamp, T. Krigas, Nature 205 (1965) 698.
[4] B. Rosenberg, L. Vancamp, J.E. Trosko, V.H. Mansour, Nature 222 (1969) 385.
3.6.8. Statistics analysis [5] J.J. Bonire, S.P. Fricker, J. Inorg. Biochem. 83 (2001) 217.
The biological results have been expressed as mean ± SEM. [6] D. Gaynora, D.M. Griffith, Dalton Trans. 41 (2012) 13239.
[7] M. Gielen (Ed.), Metal-Based Anti-Tumour Drugs, vol. 1, Fruend Publishing
Student’s t-test has been performed for the biological results and House Ltd, England, 1988.
the error has been found to be statistically insignificant at p < 0.05. [8] T. Storr, K.H. Thompson, C. Orvig, Chem. Soc. Rev. 35 (2006) 534.

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034
M. Nath et al. / Inorganica Chimica Acta xxx (2014) xxx–xxx 13

[9] A.J. Crowe, The antitumour activity of tin compounds, in: M. Gielen (Ed.), [42] V.I. Bregadze, S.A. Glazun, P.V. Petrovskii, Z.A. Starikova, V.Y. Rochev, H. Dalil,
Metal-Based Anti-Tumour Drugs, vol. 1, Fruend Publishing House Ltd, England, M. Biesemans, R. Willem, M. Gielen, D. de Vos, Appl. Organomet. Chem. 17
1988, p. 103. (2003) 453.
[10] M. Gielen, E.R.T. Tiekink, Tin compounds and their therapeutic potential, in: M. [43] K.H. Thompson, C. Orvig, Science 300 (2003) 936.
Gielen, E.R.T. Tiekink (Eds.), Metallotherapeutic Drugs and Metal-Based [44] M. Hanif, S.M. Meier, W. Kandioller, A. Bytzek, M. Hejl, C.G. Hartinger, A.A.
Diagnostic Agents: The Use of Metals in Medicine, John Wiley & Sons, Ltd, Nazarov, V.B. Arion, M.A. Jakupec, P.J. Dyson, B.K. Keppler, J. Inorg. Biochem.
2005, p. 421. 105 (2011) 224.
[11] M. Gielen, Appl. Organomet. Chem. 16 (2002) 481. and references therein. [45] <http://www.mybooch.com/2.html>, (accessed on April, 2012).
[12] (a) M. Gielen, Coord. Chem. Rev. 151 (1996) 41. and references therein; [46] (a) M.B. Taylor, Cosmet. Dermatol. 21 (1999) 26;
(b) M. Kemmer, M. Gielen, M. Biesemans, Met.-Based Drugs 5 (1998) 189. (b) P.L. Putten, Antonie Van Leeuwenhoek 45 (1979) 622.
[13] S.K. Hadjikakou, N. Hadjiliadis, Coord. Chem. Rev. 253 (2009) 235. and [47] (a) B.H. Kroes, A.J. van den Berg, Q.H.C. van Ufford, H. van Dijk, R.P. Labadie,
references therein. Planta Med. 58 (1992) 499;
[14] M.N. Xanthopoulou, S.K. Hadjikakou, N. Hadjiliadis, M. Schurmann, K. (b) M. Inoue, R. Suzuki, N. Sakaguchi, Z. Li, T. Takeda, Y. Ogihara, B.Y. Jiang, Y.
Jurkschat, A. Michaelides, S. Skoulika, T. Bakas, J.J. Binolis, S. Karkabounas, K. Chen, Biol. Pharm. Bull. 8 (1995) 1526;
Charalabopoulos, J. Inorg. Biochem. 96 (2003) 425. (c) <http://www.phytochemicals.info/phytochemicals/gallic-acid.php>,
[15] M.N. Xanthopoulou, S.K. Hadjikakou, N. Hadjiliadis, E.R. Milaeva, J.A. Gracheva, Phytochemicals, (accessed on April, 2012).
V.-Y. Tyurin, N. Kourkoumelis, K.C. Christoforidis, A.K. Metsios, S. Karkabounas, [48] H.E. Guard, W.M. Coleman III, M.M. Ross, Carbohydrate Res. 235 (1992) 41.
K. Charalabopoulos, Eur. J. Med. Chem. 43 (2008) 327. [49] L. Antonin, H. Jaroslav, M. David, Collect. Czech. Chem. Comm. 62 (1997) 1169.
[16] F. Huber, R. Schmiedgen, M. Schurmann, R. Barbieri, G. Ruisi, A. Silvestri, Appl. [50] M. Gielen, J. Meunier-Piret, M. Biesemans, R. Willem, A.E. Khloufi, Appl.
Organomet. Chem. 11 (1997) 869. Organomet. Chem. 6 (1992) 59.
[17] (a) R. Schmiedgen, F. Huber, A. Silvestri, G. Ruisi, M. Rossi, R. Barbieri, Appl. [51] S.V. Gruener, D.V. Airapetyan, A.A. Korlyukov, A.G. Shipov, Y.I. Baukov, V.S.
Organomet. Chem. 12 (1998) 861; Petrosyan, Appl. Organomet. Chem. 24 (2010) 888.
(b) M. Cagnoli, A. Alama, F. Barbieri, F. Novelli, C. Bruzzo, F. Sparatore, [52] T.V. Sizova, N.S. Yashina, V.S. Petrosyan, A.V. Yatsenko, V.V. Chernyshev, L.A.
Anticancer Drugs 9 (1998) 603. Aslanov, J. Organomet. Chem. 453 (1993) 171.
[18] I.I. Verginadis, S. Karkabounas, Y. Simos, E. Kontargiris, S.K. Hadjikakou, A. [53] J.A. Sanchez-Alcazar, A. Khodjakov, E. Schneider, Cancer Res. 61 (2001) 1038.
Batistatou, A. Evangelou, K. Charalabopoulos, Eur. J. Pharm. Sci. 42 (2011) 253. [54] J.A. Sanchez-Alcazar, J. Ruız-Cabello, I. Hernandez-Munoz, P.S. Pobre, P. de la
[19] L. Pellerito, L. Nagy, Coord. Chem. Rev. 224 (2002) 111. and references therein. Torre, E. Siles-Rivas, I. Garcıa, O. Kaplan, M.T. Munoz-Yague, J.A. Solıs-Herruzo,
[20] V. Narayanan, M. Nasr, K.D. Paull, in: M. Gielen (Ed.), Tin-Based Antitumor J. Biol. Chem. 272 (1997) 30167.
Drugs, Springer-Verlag, Berlin, 1990, p. 201. [55] Hu S. Chen, X. Li, R. Qin, Z. Mei, Acta Pharmacol. Sin. 28 (2007) 1215.
[21] M. Nath, S. Pokharia, X. Song, G. Eng, M. Gielen, M. Kemmer, M. Biesemans, R. [56] A. Takahashi, H. Matsumoto, K. Yuki, J.I. Yasumoto, A. Kajiwara, M. Aoki, Y.
Willem, D. de Vos, Appl. Organomet. Chem. 17 (2003) 305. Furusawa, K. Ohnishi, T. Ohnishi, Int. J. Radiat. Oncol. Biol. Phys. 60 (2004) 591.
[22] M. Nath, M. Vats, P. Roy, Eur. J. Med. Chem. 59 (2013) 310. [57] (a) N.P. Singh, M.T. McCoy, R.R. Trice, E.L. Schneider, Exp. Cell Res. 175 (1988)
[23] M. Nath, M. Vats, P. Roy, Synthesis, spectral characterization and anticancer 184;
screening of triorganotin(IV) carboxylates, in: M.G. Bhowon, S. J-Laullo, H.L.K. (b) P.L. Olive, J.P. Banáth, Nat. Protoc. 1 (2006) 23.
Wah, P. Ramasami (Eds.), Chemistry for Sustainable Development, ICPAC- [58] R.R. Tice, E. Agurell, D. Anderson, B. Burlinson, A. Hartmann, H. Kobayashi, Y.
Mauritius, Springer Science, 2011, p. 301. Miyamae, E. Rojas, J.C. Ryu, Y.F. Sasaki, Environ. Mol. Mutagen. 35 (2000) 206.
[24] S. Tabassum, C. Pettinari, J. Organomet. Chem. 691 (2006) 1761. [59] A. Szorcsik, L. Nagy, B. Gyurcsik, G. Vankó, R. Krämer, A. Vértes, T. Yamaguchi,
[25] C. Pellerito, P. D’Agati, T. Fiore, C. Mansueto, V. Mansueto, G. Stocco, L. Nagy, L. K. Yoshida, J. Radioanal. Nuc. Chem. 260 (2004) 459.
Pellerito, J. Inorg. Biochem. 99 (2005) 1294. [60] G.B. Deacon, R.J. Phillips, Coord. Chem. Rev. 33 (1980) 227. and references
[26] F. Cima, L. Ballarin, Appl. Organomet. Chem. 13 (1999) 697. therein.
[27] C. Pettinari, F. Marchetti, Chemical and biomedical developments in organotin [61] T.P. Lockhart, W.F. Manders, Inorg. Chem. 25 (1986) 892.
cancer chemotherapy, in: A.G. Davies, M. Gielen, K.H. Pannell, E.R.T. Tiekink [62] T.P. Lockhart, W.F. Manders, J.J. Zuckerman, J. Am. Chem. Soc. 107 (1985) 4547.
(Eds.), Tin Chemistry, Fundamentals, Frontiers, and Applications, John Wiley & [63] A. Lycka, A. Ruzicka, J. Vanecek, L. Ceslová, J. Organomet. Chem. 695 (2010)
Sons, Ltd, Chichester, 2008, p. 454. 2493.
[28] P.J. Blower, Annu. Rep. Prog. Chem. Sect. A 100 (2004) 633. [64] M. Yuan, T. Kaneko, Y. Yokoyama, M. Tsuchiya, Anal. Sci. 17 (2001) 1405.
[29] V. Chandrasekhar, P. Singh, K. Gopal, Organotin carboxylate and sulfonate [65] T.B.S. Baul, K.S. Singh, M. Holcapek, R. Jirasko, E. Rivarola, A. Linden, J.
clusters, in: A.G. Davies, M. Gielen, K.H. Pannell, E.R.T. Tiekink (Eds.), Tin Organomet. Chem. 690 (2005) 4232 (and references cited therein).
Chemistry, Fundamentals, Frontiers, and Applications, John Wiley & Sons, Ltd, [66] (a) A. Itharat, P.J. Houghton, E. Eno-Amooquaye, P.J. Burke, J.H. Sampson, A.
Chichester, 2008, p. 93. Raman, J. Ethnopharmacol. 90 (2004) 33;
[30] V. Chandrasekhar, T. Ramalingam, Organometallics 28 (2009) 2096. (b) W. Mahavorasirikul, V. Viyanant, W. Chaijaroenkul, A. Itharat, K. Na-
[31] J.H. Zhang, R.F. Zhang, C.L. Ma, D.Q. Wang, H.Z. Wang, Polyhedron 30 (2011) Bangchang, BMC Complement. Altern. Med. 10 (2010) 55;
624. (c) M.J. Moshi, E. Innocent, J.J. Magadula, D.F. Otieno, A. Weisheit, P.K. Mbabazi,
[32] M.N. Xanthopoulou, N. Kourkoumelis, S.K. Hadjikakou, N. Hadjiliadis, M. R.S.O. Nondo, Tanz. J. Health Res. 12 (2010) 63.
Kubicki, S. Karkabounas, T. Bakas, Polyhedron 27 (2008) 3318. [67] N. Nigam, K. Bhui, S. Prasad, J. George, Y. Shukla, Chem. Biol. Interact. 181
[33] C.L. Ma, Q.F. Zhang, R.F. Zhang, Inorg. Chem. 47 (2008) 7060. (2009) 77.
[34] C. Liu, S. Liu, D. Du, D. Zhu, L. Xu, J. Mol. Struct. 1003 (2011) 134. and references [68] F. Thayyullathil, S. Chathoth, A. Hago, M. Patel, S. Galadari, Biol. Med. 45 (2008)
therein. 1403.
[35] M.I. Khan, M.K. Baloch, M. Ashfaq, G. Stoter, J. Organomet. Chem. 691 (2006) [69] J.K. Yao, S. Leonard, R. Reddy, Dis. Markers 22 (2006) 83.
2254. [70] N.C.N. Tam, S. Ghatak, S.M. Ho, Prostate 55 (2003) 1.
[36] M.K. Rauf, M.A. Saeed, Imtiaz-ud-Din M. Bolte, A. Badshah, B. Mirza, J. [71] K. Kannan, S.K. Jain, Pathophysiology 7 (2000) 153.
Organomet. Chem. 693 (2008) 3043. [72] D.H. Han, M.J. Lee, J.H. Kim, Anticancer Res. 26 (2006) 3601.
[37] M. Gielen, M. Biesemans, R. Willem, Appl. Organomet. Chem. 19 (2005) 440; [73] W.L. Lin, C.J. Wang, Y.Y. Tsai, C.L. Liu, J.M. Hwang, T.H. Tseng, Arch. Toxicol. 74
M. Gielen, M. Biesemans, D. de Vos, R. Willem, J. Inorg. Biochem. 79 (2000) 139. (2000) 467.
[38] G.N. Kalud-erović, R. Paschke, S. Prashar, S. Gomez-Ruiz, J. Organomet. Chem. [74] A. Gennari, B. Vivani, C.L. Galli, M. Marinovich, R. Pieters, E. Corsini, Toxicol.
695 (2010) 1883. Appl. Pharmacol. 169 (2000) 185.
[39] S. Gomez-Ruiz, G.N. Kalud-erović, S. Prashar, E. Hey-Hawkins, A. Erić, Z. Zizak, [75] P. Bernardi, L. Scorrano, R. Colonna, V. Petronilli, F. Di-Lisa, Eur. J. Biochem. 264
Z.D. Juranić, J. Inorg. Biochem. 102 (2008) 2087. (1999) 687.
[40] C. Pettina, M. Pellei, F. Marchetti, C. Santina, M. Miliani, Polyhedron 17 (1998) [76] A. Maillaux, K. Grenet, A. Bruneel, B. Beneteau-Burnat, M. Vaubourdolle, B.
561. Baudin, Eur. J. Cell Biol. 80 (2001) 442.
[41] M. Kemmer, H. Dalil, M. Biesemans, J.C. Martins, B. Mahieu, E. Horn, D. de Vos, [77] M.S. Bentle, E.K. Reinicke, E.A. Bey, D.R. Spitz, D.A. Boothman, J. Biol. Chem. 281
E.R.T. Tiekink, R. Willem, M. Gielen, J. Organomet. Chem. 608 (2000) 63. (2006) 33684.

Please cite this article in press as: M. Nath et al., Inorg. Chim. Acta (2014), http://dx.doi.org/10.1016/j.ica.2014.02.034

You might also like