You are on page 1of 8

European Journal of Medicinal Chemistry 66 (2013) 372e379

Contents lists available at SciVerse ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Short communication

Design and synthesis of novel 4-(4-oxo-2-arylthiazolidin-3-yl)


benzenesulfonamides as selective inhibitors of carbonic anhydrase IX
over I and II with potential anticancer activity
Sharad Kumar Suthar a, *, Sumit Bansal a, Sandeep Lohan a, Vikarm Modak b,
Anil Chaudhary c, Amit Tiwari d
a
Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal 576104, Karnataka, India
b
Department of Pharmaceutical Chemistry, Maharashtra Institute of Pharmacy, Pune 411038, India
c
Mahakal Institute of Pharmaceutical Studies, Ujjain 456664, India
d
Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal 576104, India

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

Article history: The novel 4-(4-oxo-2-arylthiazolidin-3-yl)benzenesulfonamide derivatives were designed and synthe-
Received 9 December 2012 sized for selective carbonic anhydrase IX (CA IX) inhibitory activity with anticancer potential. In the CA
Received in revised form inhibition assay, 3f was found to be the most potent and selective inhibitor of CA IX with inhibitory
31 May 2013
constant (KI) value of 2.2 nM. Among the synthesized compounds, 3f showed IC50 values of 5.03 mg/ml
Accepted 2 June 2013
Available online 12 June 2013
(cisplatin: 6.56 mg/ml), 5.81 mg/ml (cisplatin: 5.85 mg/ml), and 23.93 mg/ml (cisplatin: 2.75 mg/ml) against
COLO-205, MDA-MB-231, and DU-145 cell lines, respectively. At IC50, 3f caused cell shrinkage, nuclear
condensation, and nuclear fragmentation events characteristic to apoptosis in the Hoechst 33258 and
Keywords:
Benzenesulfonamide
acridine orange-ethidium bromide staining studies of COLO-205 cells. In the Dalton’s lymphoma ascites
Thiazolidin-4-one (DLA) solid tumor model 3f decreased tumor volume by 64.83% (cisplatin: 71.62%), while increase in
Carbonic anhydrase IX mean body weight was found to be only 4.09% (cisplatin: 3.47%).
Anticancer Ó 2013 Elsevier Masson SAS. All rights reserved.
Cell lines
Daltion’s lymphoma ascites

1. Introduction in several tumor tissues [9]. The CAs, specifically isozymes CA IX


and CA XII regulate pH system by maintaining intracellular pH
Cancer is a disease of complex etiology and its chemotherapy while acidifying the extracellular pH compatible for cancer cell
requires combinations of drugs which can act simultaneously on survival and proliferation [10]. The hypoxic condition in solid tu-
different targets [1]. Therefore, development of multifunctional and mors induces expression of CA IX [9]. Chronic hypoxia leads to an
multitargeting chemotherapeutic agents is an alternative to drug increased rate of glycolysis through stabilization of hypoxia-
combination protocols [2,3]. Sulfonamides have been reported to inducible transcription factor-1 (HIF-1) [11] that results into
possess antibacterial [4], antifungal [5], insulin releasing [6], and elevated lactic acid production and drop in the intracellular pH [12].
carbonic anhydrase inhibitory activity [7]. Acetazolamide (Fig. 1), a The CAs sense and maintain the acidic tumor environment; a
sulfonamide is a potent carbonic anhydrase (CA) inhibitor. In the fundamental character of solid tumors [13] by catalyzing the
year1992, Pastoreková et al. reported the involvement of CA IX in reversible hydration of tumor cell-generated CO2 into bicarbonate
cancer [8]. Later, CA XII was also found to be coexpressed with CA IX anion (HCO þ þ
3 ) and a proton (H ) [14] and then trap H extracellu-
larly to lower down the pH [12,15]. Thus, targeting the tumor-
associated isozymes with specific inhibitors is a promising strat-
Abbreviations: CA, carbonic anhydrase; HIF-1, hypoxia-inducible transcription egy in the cancer therapy [16,17].
factor-1; DLA, Dalton’s lymphoma ascites; DCC, N,N-dicyclohexylcarbodiimide; Thiazolidin-4-ones have been reported to possess anticancer
MTT, (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; AO, acridine [18], antibacterial [19], antifungal [20], anticonvulsant [21], anti-
orange; EB, ethidium bromide; OECD, Organisation for Economic Cooperation and
Development.
tuberculosis [22], and anti-human immunodeficiency virus type 1
* Corresponding author. Tel.: þ91 8894861073; fax: þ91 820 2571998. (HIV1) [23] activities. Gududuru et al. reported that thiazolidin-4-
E-mail address: sharadpune_2009@yahoo.com (S.K. Suthar). one analogs might act as inducers of apoptosis and selectively kill

0223-5234/$ e see front matter Ó 2013 Elsevier Masson SAS. All rights reserved.
http://dx.doi.org/10.1016/j.ejmech.2013.06.003
S.K. Suthar et al. / European Journal of Medicinal Chemistry 66 (2013) 372e379 373

reaction was monitored on pre-coated Merck TLC plates using


solvent system acetone:chloroform (6:4). The synthesized com-
pounds were purified by column chromatography using silica gel
100e200 mesh and final yield of products ranged from 55% to 86%.
Formation of all newly synthesized products was confirmed by FT-
IR, NMR, and Mass spectral studies.
The FT-IR spectra of all the synthesized compounds showed
characteristic absorption bands for NeH, C]O, O]S]O, C]C, Ce
N, and CeS groups. The amino group present in sulfonamide moiety
was absorbed between 3318.78 and 3325.39 cm1. The synthesis of
thiazolidin-4-one moiety was evident by presence of vibrational
frequency ranged from 1689.70 cm1 to 1716.70 cm1 character-
istic to 4-C]O group. In all the synthesized compounds O]S]O
bonds of sulfonamide group appeared between 1333.60e1338.77
(asym.) and 1156.20e1159.45 (sym.) cm1. Bands corresponding to
C]C, CeN, and CeS groups were also observed in the FT-IR spectra.
The NMR spectra of all newly synthesized compounds showed
singlet peak for two NeH protons of sulfonamide group between
Fig. 1. Pharmacophoric similarity of acetazolamide (potent carbonic anhydrase in- 12.042 and 12.051 ppm. Further, presence of two methylene pro-
hibitor) and 2-aryl-4-oxo-thiazolidine amide (potent cytotoxic agent) with newly
synthesized compound 3f.
tons (eCH2e, 5-Ha and 5-Hb protons) indicates cyclization into
thiazolidin-4-one ring. The 5-Ha proton due to its proximity with
carbonyl group appeared downfield to 5-Hb proton either as a
a variety of prostate cancer cell lines [24]. They used thiazolidin-4- double doublet or as a doublet ranging between 4.033 and
one moiety as a biomimetic replacement for the phosphate 4.112 ppm (J1 ¼ 15.2e16 Hz and J2 ¼ 0.8 or 1.2 Hz). The 5-Hb proton
group and synthesized 2-aryl-4-oxo-thiazolidine amide based 2- appeared between 3.848 and 3.940 ppm as a doublet with J value
arylthiazolidine-4-carboxylic acid amides. The lead compound 15.6e16.4 Hz. The 2-H proton of thiazolidin-4-one ring was
(Fig. 1) obtained in their study showed IC50 values of 12.6, 11.1, 9.3, observed as a singlet between 6.631 and 6.677 ppm. All the aro-
7.1, and 8.5 mM against DU-145, PC-3, LNCaP, PPC-1, and TSU human matic protons appeared between 7.252 and 7.836 ppm. The mo-
prostate cancer cell lines, respectively [24]. Zhou et al. synthesized lecular ion peak (Mþ) corresponding to molecular weight was also
thiazolidin-4-one derivatives targeting drug-resistant lung cancer observed in GCeMS spectra of all the compounds.
cells [25]. These compounds selectively killed both non-small cell
lung cancer cell line H460 and its paclitaxel-resistant variant 2.2. Biological screening
H460taxR at an IC50 between 0.21 and 2.93 mM [25]. Based on
above findings, it was of interest to design and synthesize novel 2.2.1. CA enzyme inhibition studies
hybrid compounds possessing both sulfonamide and thiazolidin-4- All the synthesized compounds (3aej) were assayed for their
one pharmacophores (Fig. 1) and screen them for selective CA IX potential to inhibit physiologically relevant enzymes ubiquitous CA
inhibitory activity with anticancer potential. I and II, and the tumor-associated isoform CA IX by carrying out
stopped-flow assay [27]. Clinically used CA inhibitor acetazolamide
2. Results and discussion was used as reference drug. The results of CA inhibition assay
presented in Tables 1 and 2 indicated that: (i) Unlike the compound
2.1. Chemistry 3a, which exhibited potent inhibitory activity (KI of 112.3 nM)
against the slow cytosolic isoform CA I, the congeners 3bej showed
All the synthesized compounds were prepared by one pot re- moderate to weak CA I inhibitory activity with KI in the range of
action of sulfanilamide (1) with various aldehydes (2aej) and thi- 736.5e1646.6 nM. It indicated that, substitution of halogen atoms
oglycolic acid in presence of dicyclohexylcarbodiimide (DCC) to in phenyl ring of benzenesulfonamide derivatives resulted into the
yield a new series of benzenesulfonamides, containing thiazolidin- dramatic loss of CA I inhibition activity, (ii) All the compounds
4-one moiety (3aej) (Scheme 1) [26]. In the initial step amine re- showed a better activity profile against the CA II than that of CA I,
acts with aldehyde to form imine, followed by attack of sulfur (iii) All the tested derivatives exhibited significant CA IX inhibitory
nucleophile (sulfur atom of thioglycolic acid) and subsequent
intramolecular cyclization with elimination of water yields 4-(4-
oxo-2-arylthiazolidin-3-yl)benzenesulfonamides. Progress of the Table 1
Carbonic anhydrase inhibitory activity of test compounds and reference inhibitor.

Compound KI (nM)  SD

CA I CA II CA IX

3a 112.3  2.2 56.7  2.6 21.2  0.34


3b 1036.7  5.5 35.4  1.5 44.4  0.62
3c 1646.6  6.6 45.2  1.9 53.1  0.21
3d 1239.2  6.9 33.1  2.3 37.4  0.15
3e 736.5  7.5 42.6  3.8 7.7  0.14
3f 838.3  4.4 135.5  3.5 2.2  0.25
3g 742.3  6.3 38.7  2.6 4.7  0.04
3h 936.4  3.3 31.8  2.1 9.2  0.92
3i 1247.9  5.9 121.6  3.9 19.4  0.61
3j 1112.5  6.7 28.4  2.1 13.5  0.52
Acetazolamide 252.2  5.2 12.5  1.1 25.2  1.6
Scheme 1. Synthesis of 4-(4-oxo-2-arylthiazolidin-3-yl)benzenesulfonamides (3aej).
374 S.K. Suthar et al. / European Journal of Medicinal Chemistry 66 (2013) 372e379

Table 2 205, MDA-MB-231, and DU-145 cell lines, respectively. Results


Selectivity ratios of test compounds and reference CA inhibitor for the inhibition of (Table 3) indicate that halogen substitutions on aryl ring were
tumor-associated (CA IX) over the cytosolic (CA I and II) isozymes.
fruitful and resulted into increased activity. Chloro substituted
Compound Selectivity ratio compounds were more active than bromo substituted ones, while
CA I/CA IX CA II/CA IX fluoro substituted derivatives were least active. Incorporation of
halogen atom at m-position in the derivatives conferred highest
3a 5.3 2.7
3b 23.3 0.79 activity [3f (3-Cl) > 3i (3-Br) > 3c (3-F)]. The activity pattern among
3c 31.0 0.85 the synthesized compounds was followed as: m-substitution > p-
3d 33.1 0.89 substitution > o-substitution. From the above study it can be
3e 95.7 5.5
inferred that: (i) m-substitution was the most favorable for activity,
3f 381.0 61.6
3g 157.9 8.23 (ii) Substitution of chloro group imparted maximum activity, (iii)
3h 101.8 3.45 All the compounds were moderately potent or potent and no wide
3i 109.5 6.3 range of activity variation was seen; this can be attributed to the
3j 82.4 2.1 presence of common pharmacophores sulfonamide and thiazoli-
Acetazolamide 10 0.48
din-4-one in all the compounds.

2.2.3. Hoechst 33258 DNA staining to study nuclear morphological


activity with KI values between 2.2 nM and 53.1 nM, while acet- changes [29]
azolamide exhibited KI of 25.2 nM. All the benzenesulfonamide DNA-binding dye Hoechst 33258 was used to study nuclear
derivatives with halogen groups (3bej) showed greater CA IX morphological changes associated with cell death triggered by 3f in
inhibitory potential than that of compound 3a not possessing the COLO-205 cells. This dye is a fluorescent stain used to label DNA and
halogen group. The compound 3f bearing chloro group at m-posi- hence, it is commonly used to visualize nuclei. Hoechst 33258 is
tion of phenyl ring emerged as the most potent inhibitor of CA IX excited by ultraviolet light at 340e380 nm and emits blueecyan
with KI value of 2.2 nM. Among all the halogenated derivatives, fluorescence at emission maximum of 465 nm. This dye binds to the
fluoro substituted ones were found to be least potent inhibitors of minor groove of double-stranded DNA, preferably to the adenine
CA IX, (iv) A very important aspect of designing CA inhibitors is and thymine rich sequences. The apoptotic cells are more perme-
their selectivity toward CA isoforms. It is evident from Table 2 that able to Hoechst 33258 than living cells, were identified based on
newly synthesized benzenesulfonamide derivatives showed the nuclear condensation, and DNA fragmentation events of
excellent selectivity ratios for inhibiting the tumor-associated iso- Hoechst 33258 stained cells in fluorescent microscope. The results
form CA IX over the cytosolic CA I and CA II. Indeed, compound 3f showed that test compound 3f potentially induced the nuclear
exhibited the selectivity ratios of 381 and 61.6 for inhibiting the condensation and fragmentation process in COLO-205 cells, which
tumor-associated isoform CA IX over CA I and CA II, respectively. is the one of the characteristics of apoptotic cells (Fig. 2). On the
Acetazolamide exhibited the selectivity ratios of only 10 and 0.48 contrary, no morphological changes were seen in the control cells.
for the inhibition of CA IX over CA I and CA II, respectively. Though,
the selectivity ratios of compounds 3b, 3c, and 3d for CA IX over the 2.2.4. Acridine orange (AO)eethidium bromide (EB) cell
CA II were between 0.79 and 0.89; indicating that these compounds morphological analysis [30]
were better inhibitors of CA II than that of CA IX. Acridine orangeeethidium bromide (AOeEB) staining is used to
study cell shrinkage, membrane blabbing, membrane-bound
2.2.2. In vitro cytotoxicity studies (MTT assay) [28] apoptotic bodies, and nuclear fragmentation of apoptotic cells. AO
All the synthesized compounds were screened for their cyto- is a cell permeable and nucleic acid selective fluorescent dye. It
toxic potential against COLO-205 (human colon adenocarcinoma exhibits excitation maximum at 502 nm, emission maximum at
cell line), MDA-MB-231 (human breast carcinoma cell line), and 525 nm (green), and when bound to DNA, it emits green fluores-
DU-145 (human prostate carcinoma cell line) cell lines. In 48 h MTT cence. EB is an also fluorescent dye used to detect nucleic acids. It
assay results (Table 3), 3f emerged as a lead compound with IC50 of emits orangeered fluorescence and has excitation maxima of 300e
5.03 mg/ml (cisplatin: 6.56 mg/ml), 5.81 mg/ml (cisplatin: 5.85 mg/ 360 nm and an emission maximum of 590 nm. Both living and dead
ml), and 23.93 mg/ml (cisplatin: 2.75 mg/ml) against COLO-205, cells take up AO and appear green if AO is intercalated into double
MDA-MB-231, and DU-145 cell lines, respectively. While 3g stranded DNA and red if AO is intercalated into single stranded
showed IC50 values of 15.37, 7.16, and 29.28 mg/ml against COLO- RNA. While EB is taken up by dead cells only and it fluoresces or-
angeered because of its intercalation into the DNA of dead cells.
Results depicted in Fig. 3 indicate that control cells did not take up
EB and appeared faint orangeered. While cells treated with 3f at
Table 3
IC50 of test compounds toward various cell lines by MTT assay method, after 48 h.
IC50 showed obvious apoptotic characters (chromatin condensation
or fragmentation) and appeared intense orangeered, as dead cells
Compound IC50 mg/ml  SD
have ruptured membrane which allowed EB to enter into the cells.
COLO-205 MDA-MB-231 DU-145 Also due to the AO uptake, control cells appeared green while 3f
3a 30.59  0.81 23.87  0.50 39.24  0.34 treated cells appeared green to intense green as apoptotic cells have
3b 34.69  0.10 25.86  0.57 38.60  0.62 much more permeable membranes. Hence, results of our study
3c 29.84  0.74 21.36  0.89 39.15  0.21 provide a morphological proof that 3f was able to induce apoptosis
3d 31.36  0.10 22.69  0.42 37.07  0.15
as a large number of apoptotic cells were observed in AOeB dual
3e 19.51  0.15 13.57  0.11 33.27  0.14
3f 5.03  0.74 5.81  0.34 23.93  0.25 staining of COLO-205 cells.
3g 15.37  0.46 7.16  0.96 29.28  0.04
3h 27.47  0.23 20.63  0.02 35.02  0.92 2.2.5. DLA-induced solid tumor studies in mice [31e34]
3i 16.92  0.12 12.39  0.83 30.14  0.61 The overexpression of CA-IX is associated with hypoxic condi-
3j 23.05  0.90 17.48  0.13 35.09  0.52
Cisplatin 6.56  0.24 5.85  0.09 2.75  0.08
tion of solid tumors. Therefore, we decided to screen 3f against DLA
induced solid tumor model in mice.
S.K. Suthar et al. / European Journal of Medicinal Chemistry 66 (2013) 372e379 375

Fig. 2. Hoechst 33258 DNA staining for the effects of 3f on nuclear morphology of COLO-205 cells. Cells were incubated with control (c) and IC50 dose of 3f for 48 h (CC: control
cells, NC: nuclear condensation, NF: nuclear fragmentation).

2.2.5.1. Effect of 3f against DLA induced solid tumor volume in mice. only 4.09% (P < 0.0001) compared to 13.79% increase observed in
In the DLA induced solid tumor model, maximum increase in tumor control group. While in cisplatin treated group, body weight was
volume was observed on day 30th in the control group. Both the increased by 3.47%.
reference drug cisplatin and test compound 3f significantly
reduced development of solid tumor in mice (Supplementary data: 2.2.6. In silico docking studies [1]
Fig. 1, Table 1). The test drug 3f at 50 mg/kg remarkably decreased All the designed compounds (3aej) were docked into the
tumor volume by 64.83% (P < 0.0001), while cisplatin caused active site of CA IX using Autodock 4.2 software. Virtual screening
reduction in tumor volume by 71.62% (P < 0.0001). results showed estimated free energy of binding of compounds
between 6.84 and 7.15 kcal/mol. Further analysis of results in-
2.2.5.2. Effect of 3f on body weight changes in DLA induced solid dicates that Asn-62, His-64, Gln-67, Gln-92, and Thr-199 amino acid
tumor in mice. An inoculation of DLA cells in mice significantly residues were commonly involved in hydrogen bonding with li-
increased the body weight in control group (Supplementary data: gands. The docking results of the compounds are represented in
Fig. 1, Table 1). Maximum increase in body weight was observed Table 2 of Supplementary data. Analysis of docked structure of 3f
on day 30th. The test compound 3f significantly inhibited raise in into CA IX active site (Figs. 4 and 5) shows that both sulfonamide
body weight and in 3f treated group, body weight was increased by and thiazolidin-4-one moieties have been well accommodated into

Fig. 3. Apoptosis analysis of COLO-205 cells treated with control (c) and IC50 dose of 3f for 48 h. Control cells take up AO and appear green, while exclude EB and appear orangee
red. Cells treated with 3f take up both AO and EB and appear intense green and intense orangeered, respectively (apoptotic or nonviable cells have more permeable or ruptured
membranes and stained by both AO and EB). Nuclear condensation (NC) and nuclear fragmentation (NF) are evident in the figure. (For interpretation of the references to color in this
figure legend, the reader is referred to the web version of this article.)
376 S.K. Suthar et al. / European Journal of Medicinal Chemistry 66 (2013) 372e379

Fig. 6. Simplified presentation of 3f docked into the active site of CA IX. Active amino
acid residues surrounding 3f and hydrogen bonds in dashed lines are depicted in
figure.

3. Conclusion

Our approach to design and synthesize single structural scaffold


containing sulfonamide and thiazolidin-4-one pharmacophores for
Fig. 4. Docking of compound 3f within the CA IX active site. Amino acid residues and potent anticancer activity has been met with success. Virtual
Zn (II) ion (grayish sphere) interact with inhibitor is evident in figure. screening studies indicated that both of the pharmacophores were
involved in interaction with CA IX and were well accommodated
binding pocket of CA IX. Enhanced cytotoxicity of 3f may be within the active site of target protein. The sulfonamide pharma-
attributed to the coordination of catalytic Zn (II) ion by anionic cophore interacted with Asn-62, His-64, and Gln-67 residues of CA
oxygen of thiazolidin-4-one (C]O/Zn2þ, 2.3  A). The carbonyl ox- IX, while thiazolidin-4-one pharmacophore interacted with target
ygen of 3f also interacts with hydroxyl group of Thr-199. The sul- protein by means of hydrogen bonding through Thr-199 residue of
fonamide moiety present in the compounds has played a crucial the target. Among the virtually screened compounds, 3f exhibited
role in the activity as this functionality is involved in hydrogen the lowest estimated free energy of binding of 7.15 kcal/mol.
bonding with Asn-62, His-64, and Gln-67 residues of the target Molecular docking analysis showed that the most potent com-
protein. In 3f, one of the hydrogens of eSO2NH2 group binds with pound 3f was able to coordinate with catalytic Zn (II) ion that may
hydroxyl oxygen of Asn-62 (NeH/HeO, 2.3  A), while oxygen be attributed to higher binding affinity of 3f toward CA IX. Among
atoms of sulfonamide group form hydrogen bonds with eNH2 of the compounds tested for CA inhibitory activity, 3f displayed the
Asn-62 (S]O/HeN, 2.7  A) and nitrogen of His-62 (3.0  A). In- lowest KI value of 2.2 nM toward CA IX and its selectivity ratio was
teractions other than hydrogen bonding including hydrophobic and found to be greater than acetazolamide with respect to inhibition of
van der Waals were also observed. The most common amino acid cancer-associated CA IX over the inhibition of ubiquitous CA I and
residues to participate in such interactions were Leu-93 and Leu- CA II. IC50 values of 3f against COLO-205 and MDA-MB-231 cells
198. A simplified picture of 3f docked into the active site of CA IX were found to be lower than cisplatin. While compound 3g showed
is depicted in Fig. 6. IC50 value close to cisplatin against MDA-MB-231 cells. All the other
compounds also exhibited moderate potent to potent activity. The
m-substitution was the most favorable for activity and substitution
of chloro group conferred potency higher than bromo and fluoro
groups. Hoechst 33258 and AOeEB staining studies showed
apoptosis mediated cell death. Potency of 3f in DLA induced solid
tumor model in mice was also comparable to cisplatin. Hence, our
assumption in the designing of anticancer scaffold that incorpora-
tion of sulfonamide moiety will cause CA IX inhibition while that of
thiazolidin-4-one moiety will act as biomimetic replacement for
the phosphate group; might have played a greater role in the
cytotoxicity and antitumor activity of 4-(4-oxo-2-arylthiazolidin-3-
yl)benzenesulfonamides, although the detail molecular mechanism
still remains to be discovered.

4. Experimental

4.1. Chemistry

All the chemicals and reagents were purchased from Spec-


trochem, Himedia, SD fine chemicals limited, Aldrich, and Sigmae
Aldrich, India and were of analytical or reagent grades. The purity of
synthesized compounds was assessed by single spot on pre-coated
silica gel plates (TLC silica gel F254, Merck, Germany). The TLC sol-
Fig. 5. Orientation of 3f in the active site of CA IX. vent system used was acetone:chloroform (6:4). IR spectra were
S.K. Suthar et al. / European Journal of Medicinal Chemistry 66 (2013) 372e379 377

recorded on a Shimadzu FTIR-8310 (Shimadzu, Japan) using po- 1H, 5-Ha), 3.875e3.915 (d, J ¼ 16 Hz, 1H, 5-Hb). GCeMS (m/z):
tassium bromide discs. 1H NMR spectra were recorded on a Bruker 368 (Mþ).
400 MHz spectrophotometer (Bruker, USA) and chemical shifts are
presented in parts per million (d). Tetramethylsilane was used as 4.2.6. 4-(2-(3-Chlorophenyl)-4-oxothiazolidin-3-yl)
internal standard in 1H NMR. Mass spectra were recorded on GCe benzenesulfonamide (3f)
MS QP 5050, (Shimadzu, Japan). Elemental analyses were per- Yield: 63%. Mp: 155e156  C. IR (KBr, cm1): 3320.84 (NeH),
formed on 2400 CHN analyzer (PerkinElmer, USA). 1694.92 (C]O), 1337.10, 1158.62 (S]O). 1H NMR (DMSO-d6, d ppm):
12.044 (s, 2H, SO2NH2), 7.312e7.829 (m, 8H, AreH), 6.659 (s, 1H, 2-
4.2. Synthesis of 4-(4-oxo-2-arylthiazolidin-3-yl) H of thiazolidinone ring), 4.035e4.075 (d, J ¼ 16 Hz, 1H, 5-Ha),
benzenesulfonamides (3aej) [26] 3.852e3.891 (d, J ¼ 15.6 Hz, 1H, 5-Hb). GCeMS (m/z): 368 (Mþ).

General method: A mixture of amine (1.0 mmol) (1) and alde- 4.2.7. 4-(2-(4-Chlorophenyl)-4-oxothiazolidin-3-yl)
hyde (1.2 mmol) (2aej) was stirred in tetrahydrofuran under ice- benzenesulfonamide (3g)
cold condition. After 5 min, thioglycolic acid (2.0 mmol) was Yield: 83%. Mp: 174e175  C. IR (KBr, cm1): 3324.40 (NeH),
added followed by addition of dicyclohexylcarbodiimide 1698.18 (C]O), 1337.72, 1159.26 (S]O). 1H NMR (DMSO-d6, d ppm):
(1.2 mmol) at 0  C and the reaction mixture was stirred for another 12.045 (s, 2H, SO2NH2), 7.329e7.832 (m, 8H, AreH), 6.656 (s, 1H, 2-
5e6 h at room temperature (RT) to complete the reaction. The H of thiazolidinone ring), 4.040e4.078 (d, J ¼ 16 Hz, 1H, 5-Ha),
dicyclohexylurea formed during the reaction was filtered off and 3.892e3.932 (d, J ¼ 16 Hz, 1H, 5-Hb). GCeMS (m/z): 368 (Mþ).
the filtrate was concentrated to dryness under reduced pressure.
Distilled water was added to the residue and extracted with eth- 4.2.8. 4-(2-(2-Bromophenyl)-4-oxothiazolidin-3-yl)
ylacetate. The organic layer was then washed with 5% aqueous benzenesulfonamide (3h)
sodium hydrogen carbonate and sodium chloride solutions. Finally, Yield: 69%. Mp: 158e159  C. IR (KBr, cm1): 3323.00 (NeH),
organic layer was dried over anhydrous sodium sulfate and evap- 1702.26 (C]O), 1336.80, 1157.89 (S]O). 1H NMR (DMSO-d6,
orated under reduced pressure to yield solid product which was d ppm): 12.047 (s, 2H, SO2NH2), 7.315e7.819 (m, 8H, AreH), 6.677
further purified by using column chromatography. (s, 1H, 2-H of thiazolidinone ring), 4.073e4.112 (d, J ¼ 15.6 Hz, 1H, 5-
Ha), 3.888e3.927 (d, J ¼ 15.6 Hz, 1H, 5-Hb). GCeMS (m/z): 413 (Mþ).
4.2.1. 4-(4-Oxo-2-phenylthiazolidin-3-yl)benzenesulfonamide (3a)
Yield: 69%. Mp: 165e166  C. IR (KBr, cm1): 3321.20 (NeH), 4.2.9. 4-(2-(3-Bromophenyl)-4-oxothiazolidin-3-yl)
1698.10 (C]O), 1333.60, 1156.20 (S]O). 1H NMR (DMSO-d6, benzenesulfonamide (3i)
d ppm): 12.050 (s, 2H, SO2NH2), 7.252e7.808 (m, 9H, AreH), 6.631 Yield: 55%. Mp: 116e117  C. IR (KBr, cm1): 3322.95 (NeH),
(s, 1H, 2-H of thiazolidinone ring), 4.033e4.072 (d, J ¼ 15.6 Hz, 1H, 1695.63 (C]O), 1338.13, 1159.45 (S]O). 1H NMR (DMSO-d6,
5-Ha), 3.884e3.924 (d, J ¼ 16 Hz, 1H, 5-Hb). GCeMS (m/z): 334 (Mþ). d ppm): 12.051 (s, 2H, SO2NH2), 7.317e7.828 (m, 8H, AreH), 6.674
(s, 1H, 2-H of thiazolidinone ring), 4.071e4.112 (dd, J ¼ 0.8, 16 Hz,
4.2.2. 4-(2-(2-Fluorophenyl)-4-oxothiazolidin-3-yl) 1H, 5-Ha), 3.886e3.925 (d, J ¼ 15.6 Hz, 1H, 5-Hb). GCeMS (m/z): 413
benzenesulfonamide (3b) (Mþ).
Yield: 82%. Mp: 167e168  C. IR (KBr, cm1): 3318.78 (NeH),
1701.08 (C]O), 1336.42, 1158.60 (S]O). 1H NMR (DMSO-d6, 4.2.10. 4-(2-(4-Bromophenyl)-4-oxothiazolidin-3-yl)
d ppm): 12.044 (s, 2H, SO2NH2), 7.287e7.820 (m, 8H, AreH), 6.660 benzenesulfonamide (3j)
(s, 1H, 2-H of thiazolidinone ring), 4.036e4.076 (d, J ¼ 16 Hz, 1H, 5- Yield: 74%. Mp: 186e187  C. IR (KBr, cm1): 3323.46 (NeH),
Ha), 3.900e3.940 (d, J ¼ 15.6 Hz, 1H, 5-Hb). GCeMS (m/z): 352 (Mþ). 1716.70 (C]O), 1334.78, 1157.33 (S]O). 1H NMR (DMSO-d6, d ppm):
12.049 (s, 2H, SO2NH2), 7.326e7.836 (m, 8H, AreH), 6.660 (s, 1H, 2-
4.2.3. 4-(2-(3-Fluorophenyl)-4-oxothiazolidin-3-yl) H of thiazolidinone ring), 4.067e4.109 (dd, J ¼ 1.2, 16 Hz, 1H, 5-Ha),
benzenesulfonamide (3c) 3.848e3.887 (d, J ¼ 15.6 Hz, 1H, 5-Hb). GCeMS (m/z): 413 (Mþ).
Yield: 65%. Mp: 104e105  C. IR (KBr, cm1): 3320.19 (NeH),
1690.11 (C]O), 1338.62, 1159.20 (S]O). 1H NMR (DMSO-d6, 4.3. Biological screening
d ppm): 12.047 (s, 2H, SO2NH2), 7.280e7.819 (m, 8H, AreH), 6.657
(s, 1H, 2-H of thiazolidinone ring), 4.037e4.075 (d, J ¼ 15.2 Hz, 1H, 4.3.1. CA enzyme inhibition assay [27]
5-Ha), 3.894e3.933 (d, J ¼ 15.6 Hz, 1H, 5-Hb). GCeMS (m/z): An Applied Photophysics stopped-flow instrument was used to
352 (Mþ). assay the inhibition of various CA isozymes (CA catalyzed CO2 hy-
dration activity). Phenol red (at a concentration of 0.2 mM) was
4.2.4. 4-(2-(4-Fluorophenyl)-4-oxothiazolidin-3-yl) used as indicator, working at the absorbance maximum of 557 nm,
benzenesulfonamide (3d) with 10 mM Hepes (pH 7.5) as buffer and 0.1 M Na2SO4 for main-
Yield: 86%. Mp: 180e181  C. IR (KBr, cm1): 3325.39 (NeH), taining constant the ionic strength. The rate of the CA-catalyzed
1689.70 (C]O), 1338.64, 1159.26 (S]O). 1H NMR (DMSO-d6, CO2 hydration reaction was monitored for a period of 10e100 s.
d ppm): 12.042 (s, 2H, SO2NH2), 7.283e7.824 (m, 8H, AreH), 6.654 The CO2 concentrations ranged from 1.7 to 17 mM for the deter-
(s, 1H, 2-H of thiazolidinone ring), 4.034e4.076 (dd, J ¼ 1.2, 16 Hz, mination of the kinetic parameters and inhibition constants. Stock
1H, 5-Ha), 3.891e3.932 (d, J ¼ 16.4 Hz, 1H, 5-Hb). GCeMS (m/z): solutions of inhibitors (10 mM) were prepared in distillededeion-
352 (Mþ). ized water with 10e20% (v/v) DMSO and dilutions up to 0.1 nM
were made thereafter with distillededeionized water. Inhibitor and
4.2.5. 4-(2-(2-Chlorophenyl)-4-oxothiazolidin-3-yl) enzyme solutions were preincubated together for 15 min at room
benzenesulfonamide (3e) temperature prior to assay, in order to allow for the formation of
Yield: 80%. Mp: 204e205  C. IR (KBr, cm1): 3322.17 (NeH), the enzyme-inhibitor complex. The experiments were done in
1692.26 (C]O), 1338.77, 1159.39 (S]O). 1H NMR (DMSO-d6, triplicate for each inhibitor concentration, and the values reported
d ppm): 12.046 (s, 2H, SO2NH2), 7.306e7.818 (m, 8H, AreH), 6.663 in the paper are the mean of such results. The IC50 values were
(s, 1H, 2-H of thiazolidinone ring), 4.038e4.079 (dd, J ¼ 1.2, 15.6 Hz, obtained by nonlinear least-squares methods using PRISM 3 and
378 S.K. Suthar et al. / European Journal of Medicinal Chemistry 66 (2013) 372e379

represented the mean from at least three different determinations. The mice were provided with standard mice food pellets and water
Enzyme concentrations in the assay system were: 9.2 nM for CA I, ad libitum.
7.6 nM for CA II, and 12 nM for CA IX.
4.3.3.2. Toxicological study. Safe dose was calculated as per OECD
4.3.2. In vitro anticancer activity test guideline 425 [35].
4.3.2.1. Cell lines. Three human carcinoma causing cell lines viz.
COLO-205, MDA-MB-231, and DU-145 were procured from National 4.3.3.3. Preparation of test solution of compounds for animal activity.
Centre for Cell Science, Pune, India. The all three cell lines were The compound 3f was tested at the dose of 50 mg/kg body weight.
cultured in the RPMI-1640 medium with 2 mM L-glutamine The cisplatin was given as 3.5 mg/kg, i.p. The solution of test
adjusted to contain 1.5 g/l sodium bicarbonate. The medium was compound 3f was prepared by suspending it in the 0.25% carbox-
supplemented with 10% FBS, 100 U/ml penicillin, and 100 mg/ml ymethyl cellulose (CMC).
streptomycin. Cells were incubated at 37  C under 5% CO2 humid-
ified atmosphere. Dalton’s lymphoma ascites (DLA) cells were 4.3.3.4. DLA-induced solid tumor studies [31e34]. Solid tumor was
maintained and propagated by serial intraperitoneal trans- induced in mice by injecting Dalton’s lymphoma ascites cells
plantation in adult male Swiss albino mice. (1  106 cells/mouse) subcutaneously into the right hind limb of
male Swiss albino mice. The animals were grouped randomly
4.3.2.2. In vitro cytotoxicity study (MTT assay) [28]. All the newly immediately after tumor inoculation as: Group A: animals treated
synthesized compounds were screened for in vitro cytotoxicity by with vehicle, Group B: animals treated with Cisplatin (3.5 mg/kg,
MTT assay method. In brief, exponentially growing COLO-205, i.p.), and Group C: animals treated with test compound 3f (50 mg/
MDA-MB-231, and DU-145 cells were seeded into 96-well plates kg, p.o). Cisplatin (single dose, 3.5 mg/kg, i.p.) was administered on
(104 cells/well in 100 ml of media) and incubated for 24 h to obtain day 3rd which served as standard drug, while test compound 3f
70e80% confluency. The compounds to be screened were dissolved was administered on days 3rd, 5th, 7th, 10th, 12th, and 14th of
into 50 ml DMSO and were further diluted with media so as to get tumor inoculation. Tumor progression was checked by measuring
DMSO concentration below 0.25%. The cells were then treated tumor volume and body weight at the different intervals of time.
with varying concentration (250, 125, 62.5, 31.25, 15.62, and Initial diameter of the right hind limb was recorded using vernier
7.81 mg/ml) of test compounds (100 ml/well). After 48 h, media caliper. From the 3rd day onwards, tumor diameter was recorded
was discarded and 100 ml MTT reagents (1 mg/ml) was added to on every third day and volume was calculated using the formula:
cell culture and then incubated for 4 h at 37  C. The control V ¼ 4/3pr21  r2, where V is volume, r1 and r2 represent the radii of
wells contained cells with media and 0.25% DMSO. Standard the tumor at two different planes. Percentage inhibition in tumor
drug cisplatin was used as a positive control. The viable cells volume was calculated by using formula: [(Tumor volume of con-
developed formazan complex which was solubilized by adding trol animal on day 30th e Tumor volume of treated animal on day
100 ml DMSO. The plates were then kept on micro-vibrator for 30th)/(Tumor volume of control animal on day 30th)]  100.
5 min. The absorbance was measured on BIOTEK EL X800- MS The animals were weighed on day 30th and the percentage increase
microtiter plate reader at 540 nm and percentage cytotoxicity was in weight was calculated using the formula: % Increase in
calculated as (control  test/control)  100. weight ¼ [(animal weight on day 30th/animal weight on day
0)  1]  100 [31e34].
4.3.2.3. Hoechst 33258 staining to study nuclear morphological
changes [29]. COLO-205 cells (105 cells/ml) were plated into 12- 4.3.3.5. Statistical analysis. The statistical analyses were performed
well plate. When cells achieved 70e80% confluency, old media by one-way ANOVA followed by Tukey’s post hoc test using
was discarded from wells and cells were treated with IC50 of 3f and GraphPad Prism 6. The results were expressed as the mean  SEM.
incubated for 48 h. After this, cells were fixed with 4% formalde- Statistical significance was set at P < 0.05 level.
hyde for 30 min at RT and then stained with Hoechst dye for 15 min
at RT. Cells were then washed thrice with PBS and examined under
4.4. Molecular docking studies [1]
a fluorescence inverted microscope at excitation of 340e380 nm
with emission of 465 nm.
The 3D structures of ligands in pdb form were prepared by
using CS ChemDraw Ultra 8.0. The 3D crystal structure of the
4.3.2.4. Acridine orangeeethidium staining [30]. Cell death analysis
catalytic domain of the tumor-associated human carbonic anhy-
was performed by AOeEB dual staining assay. COLO-205 cells
drase IX was obtained from Protein Data Bank (PDB ID: 3IAI).
(105 cells/ml) were seeded into 12-well plate and incubated for
Acetazolamide, other hetero atoms, and water molecules were
24 h. Cells were then treated with IC50 of 3f for 48 h. Cell suspension
removed and hydrogens were added to the protein for correct
was then transferred to new vial, trypsinized and centrifuged at
calculation of partial atomic charges. The AutoDock 4.2 software
129 g for 5 min. Supernatant obtained was discarded and the pellet
was used for virtual screening studies. The grid box was set at 40,
remained behind was washed with PBS and centrifuged again.
40, and 40 A (x, y, and z) with grid point spacing of 0.375 
A. The
Finally, cells were stained with 1:1 ratio of AOeEB mixture and
Lamarckian genetic algorithm (LGA) was used to search con-
examined under fluorescence microscope.
formers with lowest binding energy. The results of docking studies
are expressed as estimated free energy of binding in kcal/mol
4.3.3. In vivo anticancer activity
(Supplementary data: Table 2).
4.3.3.1. Animals. The animal study protocol was approved by the
Institutional Animal Ethics Committee (IAEC) and complied with
the NIH guidelines on handling of experimental animals. Six to Acknowledgments
eight week old Swiss albino mice weighing 29  2 g were selected
from an inbred colony maintained under the controlled conditions The authors thank Indian Institute of Science, Bangalore, India
of temperature (30  3  C), humidity (60  5%), and light (13 and for recording NMR spectra and SAIF labs of Central Drug Research
11 h of light and dark, respectively). Four animals were housed Institute, Lucknow and Panjab University, Chandigarh, India for
in each polypropylene cage containing paddy husk as bedding. carrying out NMR spectra and CHN analysis.
S.K. Suthar et al. / European Journal of Medicinal Chemistry 66 (2013) 372e379 379

Appendix A. Supplementary data oxo-thiazolidines and their 5-arylidine derivatives, Indian J. Chem. 33B
(1994) 189e192.
[19] S.G. Küçükgüzel, E.E. Oruç, S. Rollas, F. Sahin, A. Ozbek, Synthesis, character-
Supplementary data related to this article can be found at http:// isation and biological activity of novel 4-thiazolidinones, 1,3,4-oxadiazoles
dx.doi.org/10.1016/j.ejmech.2013.06.003. and some related compounds, Eur. J. Med. Chem. 37 (2002) 197e206.
[20] G. Capan, N. Ulusoy, N. Ergenc, M. Kiraz, New 6-phenylimidazo[2,1-b]thiazole
derivatives: synthesis and antifungal activity, Monatsh. Chem. 130 (1999)
References 1399e1407.
[21] N. Ergenc, G. Capan, Synthesis and anticonvulsant activity of new 4-
[1] S.M. Marques, E.A. Enyedy, C.T. Supuran, N.I. Krupenko, S.A. Krupenko, thiazolidone and 4-thiazoline derivatives, II Farmaco 49 (1994) 133e135.
M.A. Santos, Pteridine-sulfonamide conjugates as dual inhibitors of carbonic [22] L. Bukowski, M. Janowiec, Z. Zwolska-Kwiek, Z. Andrezejczyk, Some
anhydrases and dihydrofolate reductase with potential antitumor activity, reactions of 2-cyanomethylimidazo[4,5-b]pyridine with isothiocyanates.
Bioorg. Med. Chem. 18 (2010) 5081e5089. Antituberculotic activity of the obtained compounds, Pharmazie 53 (1998)
[2] R. Morphy, Z. Rankovic, Designed multiple ligands: an emerging drug dis- 373e376.
covery paradigm, J. Med. Chem. 48 (2005) 6523e6543. [23] M.L. Barreca, A. Chimirri, L. De Luca, A.M. Monforte, P. Monforte, A. Rao,
[3] G.R. Zimmermann, J. Lehar, C.T. Keith, Multi-target therapeutics: when the M. Zappala, J. Balzarini, E. De Clercq, C. Pannecouque, M. Witvrouw, Discovery
whole is greater than the sum of the parts, Drug Discov. Today 12 (2007) 34e42. of 2,3-diaryl-1,3-thiazolidin-4-ones as potent anti-HIV-1 agents, Bioorg. Med.
[4] M.S.A. El-Gaby, A.A. Atalla, A.M. Gaber, K.A. Abd Al-Wahab, Studies on ami- Chem. Lett. 11 (2001) 1793e1796.
nopyrazoles: antibacterial activity of some novel pyrazolo[1,5-a]pyrimidines [24] V. Gududuru, E. Hurh, J.T. Dalton, D.D. Miller, Discovery of 2-arylthiazolidine-
containing sulfonamido moieties, II Farmaco 55 (2000) 596e602. 4-carboxylic acid amides as a new class of cytotoxic agents for prostate
[5] M.S.A. El-Gaby, A.M. Gaber, A.A. Atalla, K.A. Abd Al-Wahab, Novel synthesis cancer, J. Med. Chem. 48 (2005) 2584e2588.
and antifungal activity of pyrrole and pyrrolo[2,3-d]pyrimidine derivatives [25] H. Zhou, S. Wu, S. Zhai, A. Liu, Y. Sun, R. Li, Y. Zhang, S. Ekins, P.W. Swaan,
containing sulfonamide moieties, II Farmaco 57 (2002) 613e617. B. Fang, B. Zhang, B. Yan, Design, synthesis, cytoselective toxicity, structuree
[6] T.H. Maran, Relations between structure and biological activity of sulfon- activity relationships, and pharmacophore of thiazolidinone derivatives tar-
amides, Annu. Rev. Pharmacol. Toxicol. 16 (1976) 309e327. geting drug-resistant lung cancer cells, J. Med. Chem. 51 (2008) 1242e1251.
[7] C.T. Supuran, A. Scozzafava, B.C. Jurca, M.A. Iiies, Carbonic anhydrase inhibitors [26] C.V. Kavitha, Basappa, S.N. Swamy, K. Mantelingu, S. Doreswamy, M.A. Sridhar,
e part 49: synthesis of substituted ureido and thioureido derivatives of aro- J.S. Prasad, K.S. Rangappa, Synthesis of new bioactive venlafaxine analogs:
matic/heterocyclic sulfonamides with increased affinities for isozyme I, Eur. J. novel thiazolidin-4-ones as antimicrobials, Bioorg. Med. Chem. 14 (2006)
Med. Chem. 33 (1998) 83e93. 2290e2299.
[8] S. Pastoreková, Z. Zavadova, M. Kostal, O. Babusikova, J. Zavada, A novel quasi- [27] R.G. Khalifah, The carbon dioxide hydration activity of carbonic anhydrase. I.
viral agent, MaTu, is a two-component system, Virology 187 (1992) 620e626. Stop-flow kinetic studies on the native human isoenzymes B and C, J. Biol.
[9] A. Thiry, C.T. Supuram, B. Masereel, J.M. Dogné, Recent developments of car- Chem. 246 (1971) 2561e2573.
bonic anhydrase inhibitors as potential anticancer drugs, J. Med. Chem. 51 [28] T. Mosmann, Rapid colorimetric assay for cellular growth and survival:
(2008) 3051e3056. application to proliferation and cytotoxicity assays, J. Immunol. Methods 65
[10] J.C. Morris, J. Chiche, C. Grellier, M. Lopez, L.F. Bornaghi, A. Maresca, (1983) 55e63.
C.T. Supuran, J. Pouysségur, S.A. Poulsen, Targeting hypoxic tumor cell [29] X.Y. Zu, Z.Y. Zhang, X.W. Zhang, M. Yoshioka, Y.N. Yang, J. Li, Anthocyanins
viability with carbohydrate-based carbonic anhydrase IX and XII inhibitors, extracted from Chinese blueberry (Vaccinium uliginosum L.) and its anticancer
J. Med. Chem. 54 (2011) 6905e6918. effects on DLD-1 and COLO 205 cells, Chin. Med. J. 123 (2010) 2714e2719.
[11] M.C. Brahimi-Horn, J. Pouysségur, HIF at a glance, J. Cell Sci. 122 (2009) [30] K.L. Ho, L.S. Yazan, N. Ismail, M. Ismail, Apoptosis and cell cycle arrest of
1055e1057. human colorectal cancer cell line HT-29 induced by vanillin, Cancer Epi-
[12] P. Swietach, R. Vaughan-Jones, A. Harris, Regulation of tumor pH and the role demiol. 33 (2009) 155e160.
of carbonic anhydrase 9, Cancer Metastasis Rev. 26 (2007) 299e310. [31] K.M. Sakthivel, N. Kannan, A. Angeline, C. Guruvayoorappan, Anticancer ac-
[13] S. Pastoreková, J. Pastorek, Cancer-related Carbonic Anhydrase Isozymes and tivity of Acacia nilotica (L.) Wild. Ex. Delile subsp. indica against Dalton’s as-
Their Inhibition, in: Carbonic Anhydrase. Its Inhibitors and Activators, CRC cetic lymphoma induced solid and ascetic tumor model, Asian Pac. J. Cancer
Press, London, 2004, pp. 255e281. Prev. 13 (2012) 3989e3995.
[14] C.T. Supuran, Carbonic anhydrases: novel therapeutic applications for in- [32] T.D. Babu, G. Kuttan, J. Padikkala, Cytotoxic and anti-tumor properties of
hibitors and activators, Nat. Rev. Drug Discov. 7 (2008) 168e181. certain taxa of umbelliferae with special reference to Centella asiatica (L.)
[15] E. Svastova, A. Hulikova, M. Rafajova, M. Zatovicova, A. Gibadulinova, A. Casini, Urban, J. Ethnopharmacol 48 (1995) 53e57.
A. Cecchi, A. Scozzafava, C.T. Supuran, J. Pastorek, S. Pastoreková, Hypoxia [33] R.S. Jayaseelan, F.P. Vijayan, M. Mathesvaran, V. Suresh, J. Padikkala, Cytotoxic
activates the capacity of tumor associated carbonic anhydrase IX to acidify and antitumor activity of methonolic extracts desmodium triangulare (retz)
extracellular pH, FEBS Lett. 577 (2004) 439e445. merr. root, Int. J. Pharm. Pharm. Sci. 4 (2012) 540e542.
[16] A. Thiry, J.M. Dogné, J.B. Masereel, C.T. Supuran, Targeting tumorassociated [34] I. Dhamija, N. Kumar, S.N. Manjula, V. Parihar, M.M. Setty, K.S.R. Pai, Pre-
carbonic anhydrase IX in cancer therapy, Trends Pharmacol. Sci. 27 (2006) liminary evaluation of in vitro cytotoxicity and in vivo antitumor activity of
566e573. Premna herbacea Roxb. in Ehrlich ascites carcinoma model and Dalton’s
[17] S. Pastoreková, J. Kopacek, J. Pastorek, Carbonic anhydrase inhibitors and the lymphoma ascites model, Exp. Toxicol. Pathol. (2011), http://dx.doi.org/
management of cancer, Curr. Top. Med. Chem. 7 (2007) 865e878. 10.1016/j.etp.2011.08.009.
[18] J.J. Bhatt, B.R. Shah, H.P. Shah, P.B. Trivedi, N.K. Undavia, N.C. Desai, Synthesis [35] OECD Guidelines for the Testing of Chemicals: 425 Acute Oral Toxicity e Up-
of anti-HIV, anticancer and antitubercular 4-oxo-thiazolidines, 2-imino-4- and-down Procedure (UDP), OECD, 2008, pp. 1e27.

You might also like