Bioorganic & Medicinal Chemistry

You might also like

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

Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry


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

Design and synthesis of 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]


thiazole conjugates as tubulin polymerization inhibitors
Siddiq Pasha Shaik a,b, M.V.P.S. Vishnuvardhan a, Faria Sultana a, A.V. Subba Rao a,b, Chandrakant Bagul d,
Debanjan Bhattacharjee c, Jeevak Sopanrao Kapure d, Nishant Jain c, Ahmed Kamal a,b,d,e,⇑
a
Medicinal Chemistry and Pharmacology Division, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500007, India
b
Academy of Scientific and Innovative Research, New Delhi 110 025, India
c
Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500007, India
d
Department of Medicinal Chemistry, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad 500 037, India
e
Catalytic Chemistry Research Chair, Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia

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

Article history: 1,2,3-Triazolo linked benzo[d]imidazo[2,1-b]thiazole conjugates (5a–v) were designed, synthesized and
Received 6 December 2016 evaluated for their cytotoxic potency against some human cancer cell lines like DU-145 (prostate),
Accepted 11 April 2017 HeLa (cervical), MCF-7 (breast) HepG2 (liver) and A549 (lung). Preliminary results revealed that some
Available online 12 April 2017
of these conjugates like 5f and 5k exhibited significant antiproliferative effect against human breast can-
cer cells (MCF-7) with IC50 values of 0.60 and 0.78 mM respectively. Flow cytometric analysis of the cell
Keywords: cycle demonstrated an increase in the percentage of cells in the G2/M phase which was further authen-
Benzo[d]imidazo[2,1-b]thiazole
ticated by elevation of cyclin B1 protein levels. Immunocytochemistry revealed loss of intact microtubule
1,2,3-Triazole
Cytotoxicity
structure in cells treated with 5f and 5k, and western blot analysis revealed that these conjugates accu-
Tubulin depolymerization mulated more tubulin in the soluble fraction. Moreover, the conjugates caused apoptosis of the cells that
Apoptosis was confirmed by mitochondrial membrane potential and Annexin V-FITC assay. Molecular docking stud-
ies indicated that these conjugates occupy the colchicine binding site of the tubulin protein.
Ó 2017 Elsevier Ltd. All rights reserved.

1. Introduction and act as spindle poisons arresting the dividing cells in G2/M
phase of the cell cycle, causing mitotic catastrophe and finally lead-
Antimitotic agents are a major class of cytotoxic drugs for the ing to apoptotic cell death.11 Therefore, there is a growing interest
treatment of cancer and tubulin is a target for numerous small nat- in identifying and developing newer molecules that could inhibit
ural and synthetic molecules that inhibit the formation of the tubulin polymerization.
mitotic spindle.1,2 Tubulin-containing structures, such as micro- Imidazole ring is an important five-membered aromatic hetero-
tubules are responsible for the formation of the mitotic spindle cycle which is widely present in natural products and synthetic
during the process of mitosis. The microtubule system of eukary- molecules. The unique structural feature of the imidazole ring is
otic cells is a critical element in a variety of essential cellular pro- that it allows to readily bind with a variety of enzymes and recep-
cesses in addition to mitotic spindle assembly, including tors in biological systems through diverse weak interactions,
determination and maintenance of cell shape, regulation of motil- thereby exhibiting broad bioactivities like antimicrobial,12 anticon-
ity, cell signaling, secretion and intracellular transport.3 The drugs vulsant,13 sodium channel blocking,14 anti-inflammatory,15 CB1
that target microtubules and bind to the tubulin binding sites receptor inverse agonist16 and anticancer.17–19 In contrast, among
either stabilize or destabilize the microtubule assembly. There the benzoheterocycles, benzothiazole has a considerable place in
are four main binding sites of tubulin4 which includes taxane the synthetic as well as in pharmaceutical chemistry because of
and laulimalide/pelouside A sites for the microtubule stabilizing its potent and significant pharmacological activities including
agents,5–7 vinca8 and colchicine domains9,10 for the destabilizing antimicrobial,20 anti-inflammatory,21 immunosuppressive,22
agents. They interfere with the dynamic stability of microtubules antiallergenic23 and anticancer.24,25 Nitrogen-bridge head fused
heterocycles containing an imidazole and benzothiazole ring are
a common structural moiety in many pharmacologically important
⇑ Corresponding author at: Medicinal Chemistry and Pharmacology Division,
CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500007, India.
molecules that display a wide range of activities for diverse targets.
E-mail address: ahmedkamal@iict.res.in (A. Kamal). Among them, benzo[d]imidazo[2,1-b]thiazole class of compounds,

http://dx.doi.org/10.1016/j.bmc.2017.04.013
0968-0896/Ó 2017 Elsevier Ltd. All rights reserved.
3286 S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297

S O S
NEt 2.HCl N
N.HCl O N
N N
O
O N O

AC220, 2 N N
YM-201627, 1 (in phase III clinical trials) H H

O S R1
S
N N N H S
N N A B N
N N N N N
N C N
O NH
D
OMe O R2
3 OMe 4 5a-v
MeO

Fig. 1. Chemical structures of some anticancer molecules: YM-201627 (1), AC220 (2), 2-phenylbenzo[d]imidazo[2,1-b]thiazole derivative (3), 1,2,3-triazolo linked imidazo
[2,1-b]thiazole conjugates (4) and 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]thiazole conjugates (5a–v).

S R1
N N S
H N N
N N N N N
N N
O NH

OMe O R2
OMe
MeO

Fig. 2. Design strategy of 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]thiazole conjugates.

2-aryl benzo[d]imidazo[2,1-b]thiazole derivative (1, YM-201627, mopyruvate (7) in 1,2-dimethoxyethane (DME) under reflux condi-
Fig. 1) was found to be potent orally active antitumor agent useful tions afforded substituted benzo[d]imidazo[2,1-b]thiazole ester
for the treatment of solid tumors.26 Another derivative (2, AC220, (8a–d), which upon sodium hydroxide (NaOH) mediated ester
Fig. 1) was found to be highly active against FMS-like tyrosine hydrolysis gave the corresponding benzo[d]imidazo[2,1-b]thiazole
kinase-3 (FLT3) and has entered phase III clinical trials.27 Recently, acids (9a–d).39 These were coupled with propargylamine
a series of Mannich bases of 2-arylbenzo[d]imidazo[2,1-b] hydrochloride in the presence of N-(3-dimethylaminopropyl)-N0 -
thiazoles28 and earlier studies in this laboratory on 3-substituted ethylcarbodiimide hydrochloride (EDCI), triethyl amine (TEA) and
2-phenylbenzo[d]imidazo[2,1-b]thiazole29 (3, Fig. 1) showed 1-hydroxybenzotriazole (HOBT) in dry dimethylformamide (DMF)
significant cytotoxic activity with the ability to induce apoptosis under nitrogen atmosphere to provide the corresponding benzo
through inhibition of tubulin polymerization. Whereas, 1,2,3-tria- [d]imidazo[2,1-b]thiazole terminal alkynes (10a–d). The other sub-
zoles have attracted the medicinal chemists,30–32 due to their wide unit, benzyl azides (13a–f) were prepared from benzyl bromides
range of biological activities and high metabolic stability as well (12a–f) with sodium azide (NaN3) in DMSO.40 Finally, we obtained
as hydrogen-bonding capability with bimolecular targets.33–37 the desired 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]thiazole
Recently, we reported 1,2,3-triazolo linked imidazo[2,1-b]thiazole conjugates (5a–v) in high regioselectively by exposing benzo[d]im-
conjugates with potential cytotoxic and inhibition of tubulin poly- idazo[2,1-b]thiazole terminal alkynes (10a–d) to benzyl azides
merization activity (4, Fig. 1).38 (13a–f) in the presence of catalytic copper(II) sulfate pentahydrate
In continuation of our ongoing efforts on the benzo[d]imidazo (CuSO45H2O) and sodium ascorbate in a H2O/t-butyl alcohol
[2,1-b]thiazole scaffold as tubulin inhibitors and apart from the (t-BuOH) mixture at room temperature in good to excellent yields
encouraging results obtained by us for the 1,2,3-triazolo linked (65–90%). All the structures of these newly synthesized conjugates
imidazo[2,1-b]thiazole compounds,38 herein we report the synthe- (5a–v) were confirmed by spectral data (1H NMR, 13C NMR, MS and
sis of 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]thiazole conju- HRMS).
gates (5a–v) and the strategy of design is described in Fig. 2.
These conjugates were evaluated for their cytotoxic potential and
effect on tubulin polymerization, apart from the cell cycle analysis
2.2. Biological activity
to understand their possible mechanism of action.
2.2.1. Antiproliferative activity
2. Results and discussion MTT assay41 was performed to evaluate the cytotoxic effects of
these conjugates against selected human cancer cell lines, namely,
2.1. Chemistry prostate (DU-145), cervical (HeLa), breast (MCF-7), liver (HepG2)
and lung adenocarcinoma (A549) using doxorubicin as a reference
These 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]thiazole con- compound and the IC50 values are presented in Table 1. All the syn-
jugates (5a–v) were synthesized as depicted in Scheme 1. First we thesized conjugates (5a–v) displayed significant antiproliferative
have initiated our synthesis with the preparation of benzo[d]imi- activity against MCF-7 cell line when compared to other tested cell
dazo[2,1-b]thiazole derived alkyne motif’s. The condensation of lines in micro to submicromolar range except 5b, 5c and 5r.
substituted 2-aminobenzothiazoles (6a–d) with the ethylbro- Amongst them 5d, 5f, 5i, 5k and 5q showed impressive cytotoxic-
S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297 3287

R1 S
R1 O
S (a)
Br OEt N (b)
NH2 N
N OEt
O
6a-d 7 8a-d O

R1 S
R1 S
(c) N
N H
N N
N
OH 10a-d
O
9a-d R1 S
O 10a ; R 1=H
N
10b ; R1 =Me (f) N N N
N
10c; R 1=OMe NH
1 0d; R1 =OEt
O R2
5a-v
OH Br
N3 5a; R 1 =H,R2 =3-OH;4-OMe 5l; R1 =OMe,R 2 =3-OH;4-OMe
(d) (e) 5b ; R 1=H,R2 =3,4-diOMe 5m ; R 1=OMe,R 2 =3,4-diOMe
5c; R 1 =H,R2 =3,5-diOMe 5n; R 1=OMe,R 2 =3,5-diOMe
R2 R2 5d ; R 1=H,R2 =3,4,5-triOMe 5o ; R 1=OMe,R 2 =3,4,5-triOMe
R2 5e; R 1 =H,R2 =3-OPh 5p; R 1=OMe,R 2 =3-OPh
11a-f 12a-f
13a-f 5f ; R 1 =H,R2 =4-F 5q ; R 1=OMe,R 2 =4-F
5g; R 1=Me,R 2 =3,4-diOMe 5r; R 1=OEt,R 2=3,4-diOMe
13a ; R2 =3-OH;4-OMe
5h ; R 1=Me,R 2 =3,5-diOMe 5s ; R 1 =OEt,R2=3,5-diOMe
13b; R 2 =3,4-diOMe
5i; R1 =Me,R 2=3,4,5-triOMe 5t; R 1 =OEt,R2=3,4,5-triOMe
13c; R2 =3,5-diOMe
5j; R1 =Me,R 2=3-OPh 5u; R 1=OEt,R 2=3-OPh
13d ; R 2 =3,4,5-triOMe
5k ; R 1 =Me,R2 =4-F 5v ; R 1 =OEt,R2=4-F
13e; R2 =3-OPh
13f ; R2 =4-F

Scheme 1. Synthesis of 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]thiazole conjugates: Reagents and conditions: (a) DME, reflux, 8 h, 55–67%; (b) D, EtOH (hydrolysis: (i)
NaOH, H2O, EtOH, (ii) HCl), 70–75%; (c) EDCI, HOBt, propargylamine hydrochloride, Et3N, DMF, rt, 8 h, 75–79%; (d) PBr3, CH2Cl2, 0 °C, 30 min; (e) NaN3, DMSO, rt, 12 h; (f)
sodium ascorbate (10 mol%), CuSO45H2O (5 mol%), H2O/t-BuOH, (2: 1), rt, 5 h, 65–90%.

Table 1
IC50 valuesa (in mM) for compounds 10a–d and 5a–v in human cancer cell lines. ity with IC50 values of 1.412, 0.607, 1.061, 0.781 and 0.977 mM
respectively against MCF-7 cell line.
Compound DU-145b HeLac MCF-7d HepG2e A549f
In order to investigate the structure activity relationship (SAR),
10a 19.95 16.25 8.749 20.48 12.88 we have varied the substitution pattern of the ring A of benzo[d]
10b 25.11 14.79 10.03 27.58 11.74
imidazo[2,1-b]thiazole and benzyl ring (ring D) linked to the
10c 23.89 20.05 15.51 28.78 17.58
10d 22.19 20.74 17.48 24.23 16.31 1,2,3-triazole ring of these conjugates. The cytotoxicity data of
5a 14.77 12.41 9.246 16.54 9.549 compounds 10a–d deciphered that, by increasing the electron-
5b 29.83 15.31 10.28 31.620 14.120 donating nature on ring A of the benzo[d]imidazo[2,1-b]thiazole
5c 30.90 27.58 12.13 34.667 19.05
scaffold decreases the activity particularly againt MCF-7 cell line
5d 3.715 3.311 1.412 7.943 2.041
5e 21.69 14.79 6.180 21.88 10.96
when compared to other tested cell lines. In contrast, the benzyl
5f 2.570 1.629 0.607 7.413 1.570 ring (ring D) of these conjugates that has an electron withdrawing
5g 8.709 7.244 6.251 16.980 8.128 group like p-F (5f, 5k, 5q and 5v) as well as electron donating
5h 31.18 21.38 6.501 8.10 11.22 groups like 3,4,5-trimethoxy (5d, 5i and 5o) showed significant
5i 4.466 3.630 1.061 4.897 2.824
antiproliferative activity when compared to m-mono and disubsti-
5j 27.54 17.15 7.362 35.77 10.71
5k 4.265 2.691 0.781 6.606 1.288 tuted (3,4 and 3,5) electron donating group on these conjugates. It
5l 18.75 16.98 6.606 26.895 9.332 was observed that electron withdrawing p-F group showed slightly
5m 14.27 11.48 6.165 22.450 10.47 improved activity in comparison to electron donating group like
5n 13.59 11.74 8.120 15.031 14.79
3,4,5-trimethoxy substituted conjugates. Most of the correspond-
5o 9.772 8.542 2.818 17.370 4.897
5p 13.08 12.58 3.630 30.19 6.367
ing conjugates showed enhanced cytotoxicities compared to
5q 3.38 3.235 0.977 6.688 2.511 benzo[d]imidazo[2,1-b]thiazole alkynes (10a–d) particularly
5r 32.27 31.62 24.50 34.60 22.85 againt MCF-7 cell line except 5a, 5b, 5c and 5r. The activity of these
5s 9.935 7.762 7.244 15.963 14.93 conjugates like 5f, 5k and 5q improved almost by 10 folds than
5t 15.40 9.54 5.370 18.70 8.912
their alkyne intermediates (10a–d). Some of the 1,2,3-triazolo
5u 12.02 15.10 6.683 18.14 8.053
5v 14.41 12.58 3.311 17.310 8.317 linked benzo[d]imidazo[2,1-b]thiazole conjugates like 5d, 5f, 5i,
Doxorubicin 0.895 2.089 1.659 2.511 1.445 5k, 5o, 5q and 5v showed comparable or improved activity than
YM-20162728 – 4.30 3.59 2.02 – YM-201627.28 And 5f and 5k conjugates displayed promising cyto-
Note: toxicity (IC50 values of 0.667 and 0.781 mM), these are superior or
a
50% Inhibitory concentration and the values are average of three individual comparable to compound 3 (IC50 values of 0.778 mM). These results
experiments after 48 h of drug treatment. suggest that electron-donating substituents like H and Me on ring
b
Prostate cancer. A and electron withdrawing group like p-F and electron donating
c
Cervical cancer.
d
Breast cancer.
group like 3,4,5-trimethoxy on ring D are essential for the cytotoxic
e
Liver cancer. activity as shown in Fig. 3. Moreover from the MTT assay, the most
f
Lung cancer. cytotoxic conjugates that is 5f and 5k were taken up for further
3288 S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297

Hydrophobic region 2.2.3. Effect on cellular cyclin-B1, CDK1 and Aurora B levels by
R1 Tubulin binding region immunoblot analysis
S
N Cyclin-B1 is induced at the G2/M boundary to promote cell divi-
N N N sion. This protein is one of the important regulatory proteins of
N
Cytotoxicity order NH mitosis and accumulation of cyclin-B1 is an indication for G2/M
H>Me>OMe>OEt arrest.43 Since these compounds arrest the cells at G2/M phase,
O
Hydrogen R2 we investigated their effect on cyclin B1 protein levels. Thus, we
bonding region "F and tri OMe" gps treated the MCF-7 cells with the lead compounds 5f and 5k at
crucial for cytotoxicity 1 lM concentrations for 24 h and performed the immunoblot anal-
ysis for cyclin-B1 and tubulin as loading control. Therefore, the
Fig. 3. SAR analysis of 1,2,3-triazolo linked benzo[d]imidazo[2,1-b]thiazole
conjugates.
accumulation of cyclin-B1 levels suggests that these conjugates
demonstrate cytotoxic effect through cell cycle arrest at mitosis.
This data suggests that treatment by 5f and 5k induces a mitotic
mechanistic studies, such as tubulin polymerization inhibition, arrest, with the activation of spindle checkpoint in 24 h as shown
immunohistochemistry and apoptosis induction. In addition, these in Fig. 5. We have also analyzed Cdk1 and Aurora B kinases levels
conjugates were also investigated for molecular docking studies to in MCF-7 cells which are the key kinases involved in G2/M cell
understand their mode of binding. cycle transition in mammalian cells. Cdk1 is activated by the
cyclin-B1 and involved in the metaphase by activating motor pro-
2.2.2. Cell cycle analysis teins required for spindle separation. Similarly Aurora B kinase is
To investigate the mechanism underlying the cytotoxic activity also involved in phosphorylation of histone H3 protein and local-
of 5f and 5k, the cell cycle distribution in MCF-7 cancer cell line ization of some proteins at kinetochore which recognizes the spin-
was analyzed by flow cytometry.42 In this study MCF-7 cells were dle attachment at chromosome. It also binds to EB1 protein which
treated with 0.5 mM concentration for 48 h. The data obtained regulates the microtubule dynamics at spindle.44,45 Accumulation
clearly indicated that they show G2/M cell cycle arrest. Both 5f of Cdk1 and aurora B in MCF-7 cells treated with 5f and 5k is
and 5k showed 33.9% and 32.6% of cell accumulation in G2/M comparable with the positive control nocodazole. Therefore, the
phase respectively, whereas in control (untreated cells) 2.6% of increased levels of these two kinases in comparison to the
G2/M phase was observed (Fig. 4 and Table 2). untreated control cells (Fig. 5) demonstrate that MCF-7 cells

Fig. 4. Flow cytometric analysis in MCF-7 cancer cell lines after treatment with compounds at 0.5 mM concentrations for 48 h.

Table 2
Distribution of cells at G0/G1, S phase and G2/M phase of cell cycle.

Compound % of cells in Sub-G1phase % of cells in G1phase % of cells in S phase % of cells in G2/M phase
Control 3.2 83.2 11.2 2.6
5f 3.6 54.4 8.1 33.9
5k 2.9 54.1 9.4 32.6
S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297 3289

treated with 5f and 5k arrest the cells at G2/M phase due to the
inhibition of metaphase to anaphase transition.

2.2.4. Effect on tubulin polymerization


To investigate whether the anti-proliferative activities of these
conjugates were related to the interaction with tubulin, we studied
their effect on the tubulin polymerization in a cell-free system.
Moreover these conjugates showed significant effect on the G2/M
cell cycle arrest, hence it was considered of interest to investigate
the tubulin polymerization aspect.46,47 As tubulin subunits
heterodimerize and self-assemble to form microtubules in a time
dependent manner, we have investigated the progression of tubu-
lin polymerization by monitoring the increase in fluorescence
emission at 420 nm (excitation wavelength is 360 nm) in 384 well
plate for 30 min at 37 °C with and without the compounds at 3 mM
concentration. Conjugates 5f and 5k inhibited tubulin polymeriza-
tion by 49.9%, 52.3%, compared to the control (Fig. 6). Tubulin poly-
merization inhibition was also observed in case of the standards
like nocodazole (51.7%). Furthermore, these two potential
Fig. 5. Western blot analysis of Cyclin-B1, CDK1 and Aurora B: Treatment of MCF-7 conjugates (5f and 5k) were evaluated for their in vitro tubulin
cells with 1 mM concentrations of 5k and 5k for 24 h resulted an increase in cyclin polymerization assay at different concentrations. They showed
B1, CDK1 and Aurora B levels significantly. Tubulin was employed as loading control
and DMSO as negative control.
potent inhibition of tubulin polymerization with IC50 values 1.23
and 1.65 mM respectively compared to the control, where as

Fig. 6. Effect of compounds on the tubulin polymerization: tubulin polymerization was monitored by the increase in fluorescence at 360 nm (excitation) and 420 nm
(emission) for 30 min at 37 °C. All the compounds were included at a final concentration of 3 mM.
3290 S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297

Table 3
Inhibition of tubulin polymerization (IC50) for conjugates 5f and 5k.

S. no. Compound IC50a + S.D(mM)


1 5f 1.23 + 0.29
2 5k 1.65 + 0.99
3 Nocodazole 1.32 + 0.85
Fig. 8. MCF-7 cells were treated with 1 mM of 5k and 5f for 48 h. Nocodazole and
a
Concentration of drug to inhibit 50% of tubulin assembly. taxol were used as reference standards. Levels of tubulin were detected by Western
blot analysis.

nocodazole was used as the positive control that showed an IC50


value of 1.32 mM (Table 3). with 1 mM of 5k and 5f for 24 h. In addition, cells were treated with
nocodazole, as positive and taxol as negative controls in parallel
2.2.5. Immunohistochemistry studies on tubulin experiments. Western blot analysis revealed that the amount of
To validate the effect of these compounds on the disruption of tubulin protein in both soluble and polymerized fractions was
microtubule dynamics in the living cells, studies were carried out approximately the same in DMSO treated cells. Nocodazole treated
to examine the in situ effects of 5f and 5k on cellular microtubules. cells exhibited a shift of tubulin from the polymerized fraction into
MCF-7 cells seeded on sterile cover slips were treated with these the soluble fraction. In comparison, paclitaxel a microtubule poly-
compounds and nocodazole was employed as a standard at a con- merization agent showed more amount of tubulin in the polymer-
centration of 0.5 mM for 48 h. The confocal images depicted in Fig. 7 ized fraction. As expected the cells treated with 5k and 5f
showed that the untreated lung cancer cell line displayed the nor- significantly increased the tubulin content in the soluble fraction,
mal distribution of microtubules. However, cells treated with com- with almost all the tubulin present in the soluble fraction similar
pounds 5f and 5k showed disrupted microtubule organization as to that of the positive control. Therefore, increased tubulin in sol-
seen in Fig. 7, thus demonstrating the inhibition of tubulin poly- uble fraction of cells treated by these conjugates corroborated with
merization. The density of microtubules was pronounced at the the inhibition of tubulin assembly and arrest of cells in G2/M phase
cell periphery with disorganized central networks and the stan- as shown in Fig. 8.
dard nocodazole also showed disrupted microtubule organization.
These data suggested that both 5f and 5k acts as a mitotic inhibi- 2.2.7. Measurement of mitochondrial membrane potential (DWm)
tors by blocking cell cycle progression and metaphase by its ability Mitochondria plays an essential role in the propagation of apop-
to disrupt spindle assembly. tosis and its dysfunction within the apoptotic process, which is
often associated with loss or depolarization of mitochondrial mem-
2.2.6. Distribution of soluble versus polymerized tubulin in cells brane potential (DWm) leading to collapse of mitochondrial func-
Since inhibition of tubulin polymerization disturbs the micro- tions ensuing cell death.48 Therefore, in this study we examined
tubule dynamics, we evaluated the levels of soluble versus poly- the effect of conjugates 5f and 5k on DWm. In control cells the
merized forms of tubulin in MCF-7 cells following treatment dye concentrates in the mitochondrial matrix where it forms red

Fig. 7. Effect on microtubules and nuclear condensation: MCF-7 cells were independently treated with 5f, 5k and nocodazole at 0.5 mM concentration for 24 h. Following the
termination of the experiment, cells were fixed and stained for tubulin. DAPI was used as the counter stain and the merged images of cells stained for tubulin and DAPI are
seen.
S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297 3291

fluorescent aggregates (J-aggregates) because of the electrochemi- and 5k for 48 h at 0.5 mM concentration to examine the apoptotic
cal potential gradient. In cells treated with compounds which effect. The results suggested that 94.0% cells were live, 3.7% and
induce apoptosis, the mitochondrial membrane is depolarized thus 2.1% cells were in early and late apoptosis stage respectively and
preventing the accumulation of the JC-1 dye in the mitochondria. 0.1% cells were dead in the untreated MCF-7 cells (Fig. 10). How-
Therefore, the dye in the monomeric form is dispersed throughout ever, in the presence of 0.5 mM 5f and 5k, 13.7, 8.2% of cells were
the entire cell leading to a shift from red (J-aggregates) to green found to be live while 32.5, 36.6 and 53.6, 54.9% of cells were in
fluorescence (JC-1 monomers). Whereas JC-1 displays a red fluo- the early and late apoptosis stages and 0.2, 0.3% cells were necrotic
rescence (590 nm) with normal cells (high Dwmt), which is caused as shown in Fig. 10. Similarly treatment with etoposide (0.5 mM),
by the impulsive and local formation of aggregates that are related 8.5% of cells were live where as 36.1% and 55.0% of cells were in
with a large shift in the emission. In contrast, when the mitochon- the early and late stages of apoptosis respectively and 0.4% of cells
drial membrane is depolarized (low Dwmt), JC-1 forms monomers were necrotic. This experiment suggests that they significantly
that emit at 530 nm. In this study MCF-7 cells were treated with induce apoptosis in MCF-7 cells.
5f and 5k at 0.5 mM concentration for 48 h and stained with JC-1
dye. Further the loss of mitochondrial membrane potential was 2.3. Molecular docking studies
quantified by flow cytometry. From the data the results displayed
that there is a remarkable shift of JC-1 aggregates to JC-1 mono- To rationalize the experimental data obtained, molecular dock-
mers of the treated conjugates 5f and 5k that has increased to ing studies were performed on this series of conjugates (5f and 5k)
24.6% and 23.0% respectively. Whereas the control cells showed and to elucidate the possible binding mode on tubulin protein.
4.7% as seen in Fig. 9, thus indicating that both the conjugates Coordinates of protein structure of tubulin-colchicine were
induce apoptosis through depolarization of the mitochondrial obtained from the Protein Data Bank (PDB ID 3E22).50 Docking
membrane potential in MCF-7 cells. was carried out into the colchicine binding site of the tubulin using
AutoDock 4.2 software51 and analyzed by PYMOL software.52 As
2.2.8. Annexin-V FITC assay seen from Fig. 11A amide group of conjugate 5f established the
The apoptotic inducing ability is also investigated by Annexin V hydrogen binding interactions (red dotted lines) in which carbonyl
FITC/PI (AV/PI) dual staining assay to examine the occurrence of oxygen interacts with bAla354 and –NH interacts with bLeu248.
phosphatidylserine externalization and also to understand The benzo[d]imidazo[2,1-b]thiazole ring of the conjugate 5f buried
whether it is due to physiological apoptosis or nonspecific necro- in the hydrophobic pocket and surrounded by bLeu255, bLys241,
sis.49 In this study MCF-7 cells were treated with compounds 5f bLeu252, bLeu242, bVal348 and bAla316 amino acid residues.

Fig. 9. Effect on disruption of mitochondrial function and induction of apoptosis in cancer cells. Breast cancer cells were exposed to 5f and 5k for 48 h at 0.5 mM concentration
and stained with JC-1 dye for 20 min at room temperature.
3292 S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297

Fig. 10. Flow cytometric analysis of cells stained with annexin V and PI showing the distribution of Breast cancer cells in different phases of apoptosis. Cells were incubated
with 0.5 mM of 5f and 5k for 48 h.

The conjugate 5f interacts with the a and b tubulin interface and disrupt the microtubule organization in the breast cancer cells.
wherein the 1,2,3-triazole ring is surrounded by aSer148, aAla180, The molecular docking studies revealed that these conjugates bind
aSer178, bAsn258, bLeu255 and 4-fluorophenyl ring by aAsn101, at the colchicine site of the tubulin. The induction of apoptosis is
aTyr224 and bLeu248 amino acid residues. Similarly from associated with mitochondrial membrane potential and Annexin
Fig. 11B, the amide group of conjugate 5k established the hydrogen V FITC assay. Thus these conjugates could be considered as poten-
binding interactions like in case of conjugate 5f, in which carbonyl tial scaffolds that are useful in the development of new leads as
oxygen interacts with bAla354 and -NH interacts with bLeu248. chemotherapeutics for breast cancer.
Conjugate 5k establishes similar binding position like conjugate
5f and is surrounded by similar amino acid residues. Fig. 11C 4. Experimental section
shows overlapping of molecular docking pose of conjugate 5f (yel-
low) and 5k (red) whereas Fig. 11D shows that the proposed bind- 4.1. Chemistry
ing pose of conjugate 5f (yellow) and 5k (red). It is observed that
they share similar binding site as colchicine (brown) with different All the chemicals and reagents, which were commercially
orientation at interface of a-tubulin (blue) and b-tubulin (pink). received from Aldrich (Sigma-Aldrich, St. Louis, MO, USA), Lan-
Overall, docking investigation showed that conjugate 5f and 5k caster (Alfa Aesar, Johnson Matthey Company, Ward Hill, MA,
could interact with the colchicine binding site between the a and USA), or Spectrochem Pvt. Ltd (Mumbai, India) used directly with-
b subunits of tubulin. out further purification unless otherwise noted. The reaction
courses and product mixtures were routinely checked by TLC per-
3. Conclusion formed on silica gel glass plates containing 60 GF-254, and visual-
ized under UV irradiation and iodine. Column chromatography was
In the present investigation, a new class of 1,2,3-triazolo linked performed using Merck 60–120 mesh silica gel. Proton (1H) NMR
benzo[d]imidazo[2,1-b]thiazole conjugates were synthesized and spectra were recorded on Bruker UXNMR/XWIN-NMR (300 MHz)
evaluated for their cytotoxic activity. Among them, conjugates 5f or Inova Varian-VXRunity (400, 500 MHz) instruments. Carbon
and 5k showed significant cytotoxic activity against the human (13C) NMR spectra were recorded on a Bruker UXNMR/XWIN-
breast cancer cell line (MCF-7). The SAR provided an insight that NMR (75 MHz) instrument. Chemical shifts (d) are reported in d
could be utilized in developing improved leads based on these scaf- units, parts per million (ppm) downfield from an internal standard
folds. The flow cytometric analysis revealed that these conjugates TMS and coupling constants (J) were expressed in Hertz (Hz). Sig-
cause cell cycle arrest at G2/M phase. The presence of increased nal multiplicity patterns are designed as follows: s (singlet), d
levels of the cyclin B1 protein and tubulin in the soluble fraction (doublet), t (triplet), quartet (q) ds (double singlet), dd (double
of cells corroborate well with the tubulin polymerization inhibi- doublet), m (multiplet) and br s (broad singlet). ESI spectra were
tion. Furthermore, they effectively inhibit microtubule assembly recorded on a Micro mass Quattro LC using ESI+ software with a
S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297 3293

(A) (B)

(C) (D)

Fig. 11. (A) and (B) Interaction of the conjugate 5f and 5k with colchicines binding site of tubulin. This figure has been generated using the software PYMOL from the tubulin-
colchicine crystal structure. (C) Superimposition of conjugate 5f (yellow) and 5k (red) (D) Overlapping of conjugate 5f (yellow), 5k (red) and colchicine (brown) which shows
5f and 5k share the similar binding site of colchicine on tubulin. b-tubulin shows in pink and a-tubulin shows in blue color.

capillary voltage of 3.98 kV and an ESI mode positive ion trap residue was purified by column chromatography by using an ethyl
detector. High-resolution mass spectra (HRMS) were recorded on acetate and hexane solvent system to afford the title compounds
a QSTAR XL hybrid MS-MS mass spectrometer. Melting points were (10a–d).
determined in open glass capillary with an electrothermal melting
point apparatus and were uncorrected. 4.1.1.1. N-(Prop-2-yn-1-yl)benzo[d]imidazo[2,1-b]thiazole-2-carbox-
amide (10a). Pale yellow solid; 79% yield; mp: 217–219 °C; 1H
4.1.1. General method for the synthesis of 7-substituted-N-(prop-2-yn- NMR (300 MHz, CDCl3 + DMSO-d6) d 8.58 (s, 1H), 8.03 (s, 1H),
1-yl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamides (10a–d) 7.89 (d, J = 7.9 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.52 (t, J = 7.5 Hz,
The corresponding carboxylic acid (9a–d; 1.0 equiv) was dis- 1H), 7.42 (t, J = 7.5 Hz, 1H), 4.19 (dd, J = 5.6, 2.2 Hz, 2H), 2.45 (s,
solved in dry DMF, cooled to 0 °C (ice bath). EDCI (1.5 equiv), HOBt 1H); 13C NMR (75 MHz, DMSO-d6) d 161.20, 146.37, 131.48,
(1.2 equiv), propargylamine hydrochloride (1.2 equiv) and triethy- 129.46, 126.77, 125.86, 125.02, 115.75, 114.18, 81.48, 72.44,
lamine (1.2 equiv) were added slowly. After 10 min removed the 27.89; MS (ESI): m/z 256 [M+H]+; HRMS calcd for C13H10N3OS+
ice bath and the mixture was stirred at room temperature for 8 h [M+H]+ 256.0539, found 256.0554.
until the starting materials were consumed. Then ice cold water
was added to the reaction mixture and extracted with dichloro- 4.1.1.2. 7-Methyl-N-(prop-2-yn-1-yl)benzo[d]imidazo[2,1-b]thiazole-
methane (DCM). The combined organic extracts were washed with 2-carboxamide (10b). White solid; 75% yield; mp: 224–226 °C; 1H
aq NaHCO3 and dried with Na2SO4 and concentrated in vacuo. The NMR (500 MHz, CDCl3) d 8.28 (s, 1H), 7.56–7.48 (m, 2H), 7.33 (s,
3294 S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297

1H), 7.28 (s, 1H), 4.26 (dd, J = 5.5, 2.5 Hz, 2H), 2.48 (s, 3H), 2.26 (t, 4.1.2.3. N-((1-(3,5-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)
J = 2.5 Hz, 1H); 13C NMR (100 MHz, CDCl3) d 161.74, 147.31, benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5c). White solid;
141.27, 136.20, 130.51, 129.61, 127.59, 124.55, 113.90, 112.95, 72% yield; mp: 199–201 °C; 1H NMR (300 MHz, DMSO-d6) d 8.87
79.49, 71.61, 28.86, 21.44; MS (ESI): m/z 270 [M+H]+; HRMS calcd (s, 1H), 8.72 (t, J = 5.9 Hz, 1H), 8.15 (d, J = 7.9 Hz, 1H), 8.05 (d,
for C14H12N3OS+ [M+H]+ 270.0696, found 270.0700. J = 7.9 Hz, 1H), 7.99 (s, 1H), 7.58 (t, J = 7.7 Hz, 1H), 7.47 (t,
J = 7.7 Hz, 1H), 6.51–6.41 (m, 3H), 5.48 (s, 2H), 4.52 (d, J = 5.9 Hz,
4.1.1.3. 7-Methoxy-N-(prop-2-yn-1-yl)benzo[d]imidazo[2,1-b]thia- 2H), 3.71 (s, 6H); 13C NMR (75 MHz, DMSO-d6) d 161.33, 160.62,
zole-2-carboxamide (10c). White solid; 78% yield; mp: 215– 145.48, 141.52, 138.19, 131.51, 129.45, 126.77, 125.84, 125.04,
217 °C; 1H NMR (500 MHz, CDCl3) d 8.25 (s, 1H), 7.55 (d, 122.93, 115.57, 114.15, 106.10, 99.49, 55.20, 52.67, 34.23; MS
J = 8.8 Hz, 1H), 7.31 (t, J = 5.4 Hz, 1H), 7.22 (d, J = 2.4 Hz, 1H), 7.03 (ESI): m/z 449 [M+H]+; HRMS calcd for C22H21N6O3S [M+H]+
(dd, J = 8.9, 2.5 Hz, 1H), 4.26 (dd, J = 5.5, 2.6 Hz, 2H), 3.88 (s, 3H), 449.1390, found 449.1394.
2.26 (t, J = 2.5 Hz, 1H); 13C NMR (100 MHz, CDCl3) d 161.88,
158.05, 146.96, 141.20, 131.88, 125.88, 114.00, 113.90, 108.96, 4.1.2.4. N-((1-(3,4,5-Trimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)
79.62, 71.72, 56.12, 28.96; MS (ESI): m/z 286 [M+H]+; HRMS calcd benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5d). White solid;
for C14H12N3O2S+ [M+H]+ 286.0645, found 286.0649. 89% yield; mp: 210–212 °C; 1H NMR (300 MHz, CDCl3 + DMSO-
d6) d 8.79 (s, 1H), 8.57 (t, J = 5.7 Hz, 1H), 8.09 (d, J = 7.9 Hz, 1H),
4.1.1.4. 7-Ethoxy-N-(prop-2-yn-1-yl)benzo[d]imidazo[2,1-b]thiazole- 7.98–7.91 (m, 2H), 7.52 (t, J = 7.5 Hz, 1H), 7.43 (t, J = 7.6 Hz, 1H),
2-carboxamide (10d). White solid; 76% yield; mp: 159–161 °C; 1H 6.66 (s, 2H), 5.46 (s, 2H), 4.57 (d, J = 5.7 Hz, 2H), 3.76 (s, 6H), 3.67
NMR (300 MHz, CDCl3 + DMSO-d6) d 8.30 (s, 1H), 7.60 (d, (s, 3H); 13C NMR (75 MHz, DMSO-d6) d 161.34, 152.96, 146.29,
J = 8.9 Hz, 1H), 7.22 (d, J = 2.3 Hz, 1H), 7.03 (dd, J = 8.9, 2.4 Hz, 145.47, 141.53, 137.29, 131.45, 129.45, 126.77, 125.85, 125.04,
1H), 4.24 (dd, J = 5.6, 2.5 Hz, 2H), 4.10 (q, J = 7.0 Hz, 2H), 2.29 (t, 122.80, 115.54, 114.16, 105.74, 59.95, 55.88, 52.96, 34.25; MS
J = 2.5 Hz, 1H), 1.46 (t, J = 7.0 Hz, 3H); 13C NMR (75 MHz, DMSO- (ESI): m/z 479 [M+H]+; HRMS calcd for C23H23N6O4S [M+H]+
d6) d 161.27, 156.63, 153.47, 140.85, 130.78, 125.40, 118.04, 479.1496, found 479.1505.
113.38, 109.77, 106.20, 81.26, 72.38, 63.52, 14.75; MS (ESI): m/z
300 [M+H]+; HRMS calcd for C15H14N3O2S [M+H]+ 300.0801, found 4.1.2.5. N-((1-(3-Phenoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)benzo
300.0813. [d]imidazo[2,1-b]thiazole-2-carboxamide (5e). White solid; 85%
yield; mp: 199–201 °C; 1H NMR (500 MHz, CDCl3) d 8.30 (s, 1H),
4.1.2. General method for the synthesis of 1,2,3-triazolo linked benzo 7.72 (d, J = 7.9 Hz, 1H), 7.65 (d, J = 7.9 Hz, 2H), 7.54 (s, 1H), 7.48
[d]imidazo[2,1-b]thiazole conjugates (5a–v) (dt, J = 8.24, 1.06 Hz, 1H), 7.39 (dt, J = 8.08, 1.06 Hz, 1H), 7.35–
To a solution of 7-substituted-N-(prop-2-yn-1-yl)benzo[d]imi- 7.31 (m, 2H), 7.30–7.26 (m, 1H), 7.11 (dt, J = 7.47, 1.06 Hz, 1H),
dazo[2,1-b]thiazole-2-carboxamides (10a–d, 1.0 equiv) and benzyl 6.99 (dd, J = 8.6, 1.0 Hz, 2H), 6.94 (dt, J = 8.2, 2.2 Hz, 2H), 6.91 (d,
azides (13a–f, 1.10 equiv) in 2: 1 mixture of water and tert-butyl J = 1.7 Hz, 1H), 5.45 (s, 2H), 4.73 (d, J = 6.0 Hz, 2H); 13C NMR
alcohol, sodium ascorbate (0.1 equiv) and copper(II) sulfate (100 MHz, CDCl3) d 162.21, 158.17, 156.56, 147.49, 145.57,
(0.05 equiv) were added sequentially. The reaction was stirred at 141.83, 136.54, 131.76, 130.58, 129.99, 126.68, 125.95, 124.62,
room temperature for 5 h and TLC analysis indicated completion 123.91, 122.62, 122.33, 119.40, 118.68, 118.21, 113.88, 113.39,
of reaction. The solvent was concentrated under vacuum and the 53.96, 34.88; MS (ESI): m/z 481 [M+H]+; HRMS calcd for C26H21N6-
resulting mixture was extracted with dichloromethane (CH2Cl2), O2S [M+H]+ 481.1441, found 481.1444.
dried with Na2SO4 and concentrated in vacuo. The residue was
purified by column chromatography by using an ethyl acetate 4.1.2.6. N-((1-(4-Fluorobenzyl)-1H-1,2,3-triazol-4-yl)methyl)benzo[d]
and hexane solvent system to afford the pure title compounds imidazo[2,1-b]thiazole-2-carboxamide (5f). White solid; 78% yield;
(5a–v). mp: 237–239 °C; 1H NMR (300 MHz, DMSO-d6) d 8.87 (s, 1H),
8.71 (t, J = 6.0 Hz, 1H), 8.15 (d, J = 8.0 Hz, 1H), 8.05 (d, J = 7.9 Hz,
4.1.2.1. N-((1-(3-Hydroxy-4-methoxybenzyl)-1H-1,2,3-triazol-4-yl) 1H), 7.99 (s, 1H), 7.57 (t, J = 7.6 Hz, 1H), 7.46 (t, J = 7.7 Hz, 1H),
methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5a). White 7.39 (dd, J = 8.4, 5.6 Hz, 2H), 7.19 (t, J = 8.8 Hz, 2H), 5.55 (s, 2H),
solid; 67% yield; mp: 179–181 °C; 1H NMR (300 MHz, CDCl3 + 4.51 (d, J = 6.0 Hz, 2H); 13C NMR (75 MHz, DMSO-d6) d 161.83 (JC-
DMSO-d6) d 9.10 (s, 1H), 8.86 (s, 1H), 8.69 (t, J = 5.8 Hz, 1H), 8.14 F = 244 Hz), 161.35, 146.29, 145.54, 141.51, 132.41, 131.51,
(d, J = 7.9 Hz, 1H), 8.04 (d, J = 7.9 Hz, 1H), 7.89 (s, 1H), 7.56 (t, 130.28 (JC-F = 8 Hz), 129.44, 126.77, 125.84, 125.04, 122.83,
J = 7.6 Hz, 1H), 7.45 (t, J = 7.6 Hz, 1H), 6.87 (d, J = 8.6 Hz, 1H), 6.74 115.63 (JC-F = 6 Hz),115.39, 114.15, 51.87, 34.23; MS (ESI): m/z
(s, 2H), 5.39 (s, 2H), 4.50 (d, J = 5.8 Hz, 2H), 3.72 (s, 3H); 13C NMR 407 [M+H]+; HRMS calcd for C20H16FN6OS [M+H]+ 407.1085, found
(75 MHz, DMSO-d6) d 161.34, 147.62, 146.60, 146.32, 145.41, 407.1090.
141.51, 131.52, 129.46, 128.48, 126.77, 125.85, 125.04, 122.56,
119.08, 115.59, 115.38, 114.15, 112.22, 55.61, 52.48, 34.24; MS 4.1.2.7. N-((1-(3,4-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)-
(ESI): m/z 435 [M+H]+; HRMS calcd for C21H19N6O3S [M+H]+ 7-methylbenzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5g). White
435.1234, found 435.1235. solid; 78% yield; mp: 179–181 °C; 1H NMR (300 MHz, DMSO-d6)
d 8.79 (s, 1H), 8.67 (t, J = 5.9 Hz, 1H), 8.00 (d, J = 8.2 Hz, 1H), 7.93
4.1.2.2. N-((1-(3,4-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl) (s, 1H), 7.82 (s, 1H), 7.36 (d, J = 8.1 Hz, 1H), 7.00 (s, 1H), 6.95–
benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5b). White solid; 6.82 (m, 2H), 5.45 (s, 2H), 4.49 (d, J = 5.9 Hz, 2H), 3.71 (s, 6H),
79% yield; mp: 219–221 °C; 1H NMR (400 MHz, CDCl3) d 8.29 (s, 2.41 (s, 3H); 13C NMR (75 MHz, DMSO-d6) d 161.34, 148.70,
1H), 7.72 (d, J = 7.9 Hz, 1H), 7.65 (d, J = 7.9 Hz, 2H), 7.53–7.45 (m, 146.06, 145.44, 141.33, 135.61, 129.43, 128.25, 127.59, 124.80,
2H), 7.40 (t, J = 7.7 Hz, 1H), 6.90–6.80 (m, 2H), 6.79 (s, 1H), 5.42 122.55, 120.66, 115.43, 113.77, 112.11, 111.86, 55.48, 52.60,
(s, 2H), 4.72 (d, J = 6.0 Hz, 2H), 3.87 (s, 3H), 3.83 (s, 3H); 13C NMR 34.22, 20.88; MS (ESI): m/z 463 [M+H]+; HRMS calcd for C23H23N6-
(75 MHz, DMSO-d6) d 161.33, 148.70, 146.29, 145.43, 141.52, O3S [M+H]+ 463.1547, found 463.1548.
131.50, 129.44, 128.25, 126.76, 125.83, 125.02, 122.58, 120.67,
120.25, 115.56, 114.14, 112.11, 111.85, 55.48, 52.62, 34.24; MS 4.1.2.8. N-((1-(3,5-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)-
(ESI): m/z 449 [M+H]+; HRMS calcd for C22H21N6O3S [M+H]+ 7-methylbenzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5h). Light
449.1390, found 449.1390. brown solid; 72% yield; mp: 229–231 °C; 1H NMR (300 MHz,
S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297 3295

DMSO-d6) d 8.79 (s, 1H), 8.69 (t, J = 5.9 Hz, 1H), 8.04–7.95 (m, 2H), 479 [M+H]+; HRMS calcd for C23H23N6O4S [M+H]+ 479.1496, found
7.81 (s, 1H), 7.36 (d, J = 8.2 Hz, 1H), 6.50–6.39 (m, 3H), 5.47 (s, 2H), 479.1498.
4.51 (d, J = 5.9 Hz, 2H), 3.70 (s, 6H), 2.41 (s, 3H); 13C NMR (75 MHz,
DMSO-d6) d 161.38, 160.62, 146.07, 145.52, 141.33, 138.19, 135.60, 4.1.2.14. N-((1-(3,5-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)-
129.43, 127.58, 124.78, 122.93, 115.44, 113.77, 106.10, 99.49, 7-methoxybenzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5n).
55.19, 52.68, 34.23, 20.88; MS (ESI): m/z 463 [M+H]+; HRMS calcd Brown solid; 79% yield; mp: 208–210 °C; 1H NMR (300 MHz,
for C23H23N6O3S [M+H]+ 463.1547, found 463.1531. DMSO-d6) d 8.77 (s, 1H), 8.66 (t, J = 5.8 Hz, 1H), 8.04 (d, J = 8.9 Hz,
1H), 7.97 (s, 1H), 7.67 (d, J = 2.2 Hz, 1H), 7.14 (dd, J = 8.9, 2.3 Hz,
4.1.2.9. 7-Methyl-N-((1-(3,4,5-trimethoxybenzyl)-1H-1,2,3-triazol-4-yl) 1H), 6.46 (s, 2H), 6.44 (d, J = 1.8 Hz, 1H), 5.47 (s, 2H), 4.50 (d,
methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5i). White J = 5.8 Hz, 2H), 3.82 (s, 3H), 3.70 (s, 6H); 13C NMR (75 MHz,
solid; 85% yield; mp: 175–177 °C; 1H NMR (300 MHz, DMSO-d6) DMSO-d6) d 163.40, 161.44, 160.63, 157.38, 141.12, 138.19,
d 8.79 (s, 1H), 8.68 (t, J = 5.9 Hz, 1H), 8.02–7.99 (m, 2H), 7.82 (s, 130.81, 125.56, 122.93, 115.38, 114.78, 113.97, 109.33, 106.10,
1H), 7.36 (d, J = 8.2 Hz, 1H), 6.69 (s, 2H), 5.46 (s, 2H), 4.50 (d, 99.50, 55.87, 55.20, 52.69, 34.22; MS (ESI): m/z 501 [M+H]+; HRMS
J = 5.9 Hz, 2H), 3.73 (s, 6H), 3.61 (s, 3H), 2.41 (s, 3H); 13C NMR calcd for C23H22N6NaO4S [M+Na]+ 501.1315, found 501.1317.
(75 MHz, DMSO-d6) d 161.36, 152.96, 146.07, 145.49, 141.34,
137.31, 135.61, 131.44, 129.43, 127.58, 124.78, 122.79, 115.41, 4.1.2.15. 7-Methoxy-N-((1-(3,4,5-trimethoxybenzyl)-1H-1,2,3-triazol-
113.77, 105.73, 59.94, 55.88, 52.96, 34.23, 20.88; MS (ESI): m/z 4-yl)methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5o).
493 [M+H]+; HRMS calcd for C24H25N6O4S [M+H]+ 493.1653, found Light brown solid; 90% yield; mp: 201–203 °C; 1H NMR
493.1651. (300 MHz, DMSO-d6) d 8.76 (s, 1H), 8.64 (t, J = 5.9 Hz, 1H), 8.04
(d, J = 8.9 Hz, 1H), 7.99 (s, 1H), 7.67 (d, J = 2.3 Hz, 1H), 7.13 (dd,
4.1.2.10. 7-Methyl-N-((1-(3-phenoxybenzyl)-1H-1,2,3-triazol-4-yl) J = 8.9, 2.4 Hz, 1H), 6.69 (s, 2H), 5.46 (s, 2H), 4.50 (d, J = 5.9 Hz,
methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5j). Brown 2H), 3.82 (s, 3H), 3.72 (s, 6H), 3.61 (s, 3H); 13C NMR (75 MHz,
solid; 81% yield; mp: 132–134 °C; 1H NMR (300 MHz, DMSO-d6) DMSO-d6) d 161.41, 157.38, 152.95, 145.76, 145.49, 141.11,
d 8.81 (s, 1H), 8.70 (t, J = 5.8 Hz, 1H), 8.02–7.99 (m, 2H), 7.82 (s, 137.31, 131.43, 130.79, 125.52, 122.78, 115.33, 114.76, 113.95,
1H), 7.41–7.31 (m, 4H), 7.11 (t, J = 7.4 Hz, 1H), 7.05 (d, J = 7.6 Hz, 109.31, 105.74, 59.95, 55.88, 52.96, 34.22; MS (ESI): m/z 509 [M
1H), 6.99 (d, J = 7.6 Hz, 3H), 6.92 (d, J = 8.0 Hz, 1H), 5.55 (s, 2H), +H]+; HRMS calcd for C24H25N6O5S [M+H]+ 509.1602, found
4.51 (d, J = 5.8 Hz, 2H), 2.41 (s, 3H); 13C NMR (75 MHz, DMSO-d6) 509.1600.
d 161.38, 156.89, 156.16, 145.57, 141.33, 138.28, 135.61, 130.38,
130.06, 129.43, 127.60, 124.80, 123.65, 122.97, 122.83, 118.77, 4.1.2.16. 7-Methoxy-N-((1-(3-phenoxybenzyl)-1H-1,2,3-triazol-4-yl)
117.98, 117.88, 115.46, 113.77, 52.26, 34.23, 20.89; MS (ESI): m/z methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5p). White
495 [M+H]+; HRMS calcd for C27H23N6O2S [M+H]+ 495.1598, found solid; 81% yield; mp: 211–213 °C; 1H NMR (300 MHz, CDCl3 +
495.1592. DMSO-d6) d 8.75 (s, 1H), 8.58 (t, J = 5.9 Hz, 1H), 8.03 (d, J = 8.9 Hz,
1H), 7.97 (s, 1H), 7.62 (d, J = 2.1 Hz, 1H), 7.40–7.30 (m, 3H), 7.12
4.1.2.11. N-((1-(4-Fluorobenzyl)-1H-1,2,3-triazol-4-yl)methyl)-7-methyl- (d, J = 7.6 Hz, 2H), 7.05 (d, J = 7.8 Hz, 1H), 6.98 (d, J = 7.6 Hz, 3H),
benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5k). White solid; 6.91 (d, J = 8.4 Hz, 1H), 5.55 (s, 2H), 4.53 (d, J = 5.9 Hz, 2H), 3.84
73% yield; mp: 243–245 °C; 1H NMR (400 MHz, CDCl3) d 8.25 (s, (s, 3H); 13C NMR (100 MHz, DMSO-d6) d 161.43, 157.39, 156.89,
1H), 7.65 (s, 1H), 7.53 (s, 1H), 7.51 (s, 2H), 7.30–7.23 (m, 3H), 156.17, 145.77, 145.60, 141.11, 138.29, 130.81, 130.40, 130.08,
7.05 (t, J = 8.6 Hz, 2H), 5.46 (s, 2H), 4.72 (d, J = 6.1 Hz, 2H), 2.48 125.55, 123.67, 122.97, 122.84, 118.78, 117.98, 117.89, 115.41,
(s, 3H); 13C NMR (100 MHz, CDCl3) d 163.0 (JC-F = 247 Hz), 162.34, 114.78, 113.99, 109.35, 55.88, 52.26, 34.22; MS (ESI): m/z 511
147.44, 145.74, 141.56, 136.31, 130.64, 130.14 (JC-F = 8 Hz), [M+H]+; HRMS calcd for C27H23N6O3S [M+H]+ 511.1547, found
129.71, 127.68, 124.69, 122.23, 116.26 (JC-F = 22 Hz), 113.84, 511.1548.
113.01, 53.58, 34.87, 21.55; MS (ESI): m/z 421 [M+H]+; HRMS calcd
for C21H18FN6OS [M+H]+ 421.1241, found 421.1244. 4.1.2.17. N-((1-(4-Fluorobenzyl)-1H-1,2,3-triazol-4-yl)methyl)-7-methoxy-
benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5q). White solid;
4.1.2.12. N-((1-(3-Hydroxy-4-methoxybenzyl)-1H-1,2,3-triazol-4-yl) 75% yield; mp: 253–255 °C; 1H NMR (300 MHz, CDCl3 +
methyl)-7-methoxybenzo[d]imidazo[2,1-b]thiazole-2-carboxamide DMSO-d6) d 8.74 (s, 1H), 8.57 (t, J = 5.7 Hz, 1H), 8.02 (d, J = 8.9 Hz,
(5l). White solid; 65% yield; mp: 208–210 °C; 1H NMR (300 MHz, 1H), 7.96 (s, 1H), 7.60 (s, 1H), 7.42–7.33 (m, 2H), 7.13 (dd,
DMSO-d6) d 9.10 (s, 1H), 8.77 (s, 1H), 8.64 (t, J = 5.9 Hz, 1H), 8.04 J = 15.0, 7.4 Hz, 3H), 5.55 (s, 2H), 4.52 (d, J = 5.7 Hz, 2H), 3.84 (s,
(d, J = 8.9 Hz, 1H), 7.88 (s, 1H), 7.68 (d, J = 2.0 Hz, 1H), 7.15 (dd, 3H); 13C NMR (75 MHz, CDCl3 + DMSO-d6) d 161.43, 157.39,
J = 8.8, 2.1 Hz, 1H), 6.88 (d, J = 8.7 Hz, 1H), 6.75 (s, 2H), 5.39 (s, 145.78, 145.59, 141.10, 132.41, 130.81, 130.29 (JC-F = 8 Hz),
2H), 4.49 (d, J = 5.7 Hz, 2H), 3.83 (s, 3H), 3.73 (s, 3H); 13C NMR 125.54, 122.82, 115.69, 115.40, 114.78, 113.98, 109.34, 55.88,
(75 MHz, CDCl3 + DMSO-d6) d 161.41, 160.56, 158.39, 157.39, 51.89, 34.21; MS (ESI): m/z 437 [M+H]+; HRMS calcd for
147.62, 146.58, 130.82, 128.48, 125.55, 122.53, 119.08, 115.37, C21H18FN6O2S [M+H]+ 437.1190, found 437.1194.
114.77, 113.97, 112.23, 109.35, 55.87, 55.62, 52.47, 34.20; MS
(ESI): m/z 465 [M+H]+; HRMS calcd for C22H21N6O4S [M+H]+ 4.1.2.18. N-((1-(3,4-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)-
465.1340, found 465.1343. 7-ethoxybenzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5r). White
solid; 80% yield; mp: 214–216 °C; 1H NMR (300 MHz, DMSO-d6)
4.1.2.13. N-((1-(3,4-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)- d 8.77 (s, 1H), 8.64 (t, J = 5.8 Hz, 1H), 8.03 (d, J = 8.9 Hz, 1H), 7.93
7-methoxybenzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5m). (s, 1H), 7.67 (d, J = 2.0 Hz, 1H), 7.14 (dd, J = 8.9, 2.1 Hz, 1H), 7.00
White solid; 74% yield; mp: 199–201 °C; 1H NMR (400 MHz, CDCl3) (s, 1H), 6.93–6.85 (m, 2H), 5.46 (s, 2H), 4.49 (d, J = 5.8 Hz, 2H),
d 8.21 (s, 1H), 7.62 (s, 1H), 7.54 (d, J = 8.8 Hz, 1H), 7.50 (s, 1H), 7.21 4.09 (q, J = 6.8 Hz, 2H), 3.72 (s, 6H), 1.36 (t, J = 6.9 Hz, 3H); 13C
(s, 1H), 7.02 (d, J = 8.4 Hz, 1H), 6.89–6.80 (m, 2H), 6.78 (s, 1H), 5.42 NMR (75 MHz, DMSO-d6) d 161.41, 156.60, 148.69, 145.80,
(s, 2H), 4.72 (d, J = 5.8 Hz, 2H), 3.88 (s, 3H), 3.87 (s, 3H), 3.83 (s, 141.07, 130.75, 128.24, 125.42, 122.55, 120.66, 115.31, 114.74,
3H); 13C NMR (100 MHz, CDCl3) d 162.29, 158.03, 149.58, 146.93, 114.34, 112.11, 111.85, 109.81, 63.89, 55.48, 52.61, 34.21, 14.54;
145.54, 141.45, 131.90, 127.03, 125.88, 122.10, 121.04, 113.92, MS (ESI): m/z 493 [M+H]+; HRMS calcd for C24H25N6O4S [M+H]+
113.76, 111.44, 111.35, 108.94, 56.09, 54.26, 34.89; MS (ESI): m/z 493.1653, found 493.1651.
3296 S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297

4.1.2.19. N-((1-(3,5-Dimethoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)- 4.2.2. Cell cycle analysis


7-ethoxybenzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5s). Light Flow cytometric analysis (FACS) analysis was performed to
brown solid; 85% yield; mp: 223–225 °C; 1H NMR (500 MHz, evaluate the distribution of the cells through the cell cycle phases.
DMSO-d6) d 8.77 (s, 1H), 8.63 (t, J = 6.0 Hz, 1H), 8.03 (d, J = 8.9 Hz, Human breast cancer cells (MCF-7) were incubated with com-
1H), 7.97 (s, 1H), 7.66 (d, J = 2.3 Hz, 1H), 7.13 (dd, J = 8.9, 2.4 Hz, pounds 5f and 5k at 0.5 lM concentration for 48 h. Control and
1H), 6.46 (d, J = 1.9 Hz, 2H), 6.44 (d, J = 2.0 Hz, 1H), 5.47 (s, 2H), treated cells were harvested, washed with PBS, fixed in ice-cold
4.50 (d, J = 6.0 Hz, 2H), 4.10 (q, J = 6.9 Hz, 2H), 3.70 (s, 6H), 1.36 70% ethanol and stained with propidium iodide (Sigma Aldrich).
(t, J = 6.9 Hz, 3H); 13C NMR (75 MHz, DMSO-d6) d 161.41, 160.62, Cell cycle analysis was performed by flow cytometryR (Becton
156.61, 145.52, 141.09, 138.19, 130.76, 125.44, 122.92, 115.35, Dickinson FACS Caliber) as described earlier.42
114.75, 114.35, 109.84, 106.09, 99.49, 63.90, 55.19, 52.67, 34.20,
14.55; MS (ESI): m/z 493 [M+H]+; HRMS calcd for C24H25N6O4S
[M+H]+ 493.1653, found 493.1653. 4.2.3. Dot-blot assay
Cells were trypsinized when sub-confluent from T25
flasks/60 mm dishes and seeded in 6-well plates. These conjugates
4.1.2.20. 7-Ethoxy-N-((1-(3,4,5-trimethoxybenzyl)-1H-1,2,3-triazol- (5f and 5k) were evaluated for their activity against Cyclin B1.
4-yl)methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5t). MCF-7 cells were treated with 1 lM concentrations of 5f and 5k
White solid; 88% yield; mp: 208–209 °C; 1H NMR (300 MHz, for 24 h. Subsequently, cells were harvested and proteins were
DMSO-d6) d 8.77 (s, 1H), 8.65 (t, J = 5.9 Hz, 1H), 8.08–7.96 (m, quantified using Amido Black followed by densitometry analysis.
2H), 7.67 (d, J = 2.3 Hz, 1H), 7.14 (dd, J = 8.9, 2.4 Hz, 1H), 6.70 (s, Equal amount of protein were blotted on nitrocellulose membrane
2H), 5.46 (s, 2H), 4.50 (d, J = 5.9 Hz, 2H), 4.09 (q, J = 6.9 Hz, 2H), using Bio-Dot SF microfiltration apparatus (Bio-Rad). Briefly, nitro-
3.73 (s, 6H), 3.62 (s, 3H), 1.36 (t, J = 6.9 Hz, 3H); 13C NMR cellulose membrane and 3 filters papers (Whatmann 3) were
(101 MHz, CDCl3) d 162.32, 157.38, 153.78, 146.94, 145.66, soaked in 1 TBS solution for 10 min. Later, the filter papers, mem-
141.36, 138.38, 131.78, 130.13, 125.71, 122.29, 114.40, 113.90, brane were arranged in the apparatus and connected to vacuum
113.73, 109.49, 105.41, 64.51, 60.96, 56.31, 54.58, 34.88, 14.85; pump (Millipore). The membranes were rehydrated using 100 ll
MS (ESI): m/z 523 [M+H]+; HRMS calcd for C25H27N6O5S [M+H]+ of 1 TBS by vacuum filtration. Subsequently, 50 ll volumes of
523.1758, found 523.1764. equal protein samples were blotted on the membrane and washed
with 200 ll of 1 TBS through application of vacuum. The blot was
blocked with 5% blotto for 1 h at room temperature. Immunoblot
4.1.2.21. 7-Ethoxy-N-((1-(3-phenoxybenzyl)-1H-1,2,3-triazol-4-yl) analysis was performed using UVP, biospectrum 810 imaging
methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5u). White system.
solid; 83% yield; mp: 191–193 °C; 1H NMR (400 MHz, CDCl3) 13C
NMR (75 MHz, DMSO-d6) d 161.45, 156.90, 156.63, 156.18,
145.77, 145.59, 141.09, 138.29, 130.78, 130.40, 130.07, 125.45, 4.2.4. In vitro tubulin polymerization
123.67, 122.97, 122.84, 118.78, 117.99, 117.90, 115.37, 114.77, A fluorescence based tubulin polymerization assay was per-
114.38, 109.85, 63.93, 52.27, 34.22, 14.57; MS (ESI): m/z 525 formed according to the manufacturers protocol (BK011,
[M+H]+; HRMS calcd for C28H25N6O3S [M+H]+ 525.1703, found Cytoskeleton, Inc.). Briefly, the reaction mixture was taken within
525.1706. a total volume of 10 mL contained PEM buffer, GTP (1 mM) in the
presence or absence of test compounds 5f and 5k at a final concen-
tration of 3 mM. In vitro Tubulin polymerization was followed by a
4.1.2.22. 7-Ethoxy-N-((1-(4-fluorobenzyl)-1H-1,2,3-triazol-4-yl) time dependent increase in fluorescence due to the incorporation
methyl)benzo[d]imidazo[2,1-b]thiazole-2-carboxamide (5v). White of a fluorescence reporter into microtubules as polymerization pro-
solid; 76% yield; mp: 255–257 °C; 1H NMR (300 MHz, DMSO-d6) ceeds. Fluorescence emission at 420 nm (excitation wavelength is
d 8.78 (s, 1H), 8.65 (t, J = 6.0 Hz, 1H), 8.12–7.95 (m, 2H), 7.67 (s, 360 nm) was measured by using a Biotek multimode plate reader.
1H), 7.38 (t, J = 6.8 Hz, 2H), 7.88–7.09 (m, 3H), 5.55 (s, 2H), 4.50 Polymerization was finally monitored by increase in the fluores-
(d, J = 4.6 Hz, 2H), 4.10 (d, J = 6.8 Hz, 2H), 1.36 (t, J = 6.3 Hz, 3H); cence mentioned above at 37 °C. The IC50 value was defined as
13
C NMR (75 MHz, CDCl3 + DMSO-d6) d 163.46, 161.44, 156.63, the drug concentration required for inhibiting 50% of tubulin
145.77, 145.59, 141.08, 132.38, 130.77, 130.29 (JC-F = 8 Hz), assembly compared to the control.
125.44, 122.83, 115.69, 115.40, 114.77, 114.37, 109.85, 63.92,
51.89, 34.21, 14.56; MS (ESI): m/z 461 [M+H]+; HRMS calcd for
C22H20FN6O2S [M+H]+ 451.1347, found 451.1350. 4.2.5. Immunohistochemistry of tubulin and analysis of nuclear
morphology
MCF-7 cells were seeded on glass cover slip, incubated for 24 h
4.2. Biology in the presence or absence of test conjugates 5k and 5f (0.5 lM).
Cells grown on cover slips were fixed in 3.5% formaldehyde in
4.2.1. Anticancer activity MTT assay phosphate-buffered saline (PBS) pH 7.4 for 10 min at room temper-
The cytotoxic activity of the compounds was determined using ature. Cells were permeabilized for 6 min in PBS containing 0.5%
MTT assay. 1  104 cells/well were seeded in 200 ml DMEM, supple- Triton X-100 (Sigma) and 0.05% Tween-20 (Sigma). The permeabi-
mented with 10% FBS in each well 96 well microculture plates and lized cells were blocked with 2% BSA (Sigma) in PBS for 1 h. Later,
incubated for 24 h at 37 °C in a CO2 incubator. Compounds, diluted the cells were incubated with primary antibody for tubulin from
to the desired concentrations in culture medium, were added to (sigma) at (1:200) diluted in blocking solution for 4 h at room tem-
the wells with respective vehicle control. After 48 h of incubation, perature. Subsequently the antibodies were removed and the cells
10 mL of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra- were washed thrice with PBS. Cells were then incubated with FITC
zolium bromide) (5 mg/mL) was added to each well and the plates labeled anti-mouse secondary antibody (1:500) for 1 h at room
were further incubated for 4 h. Then the supernatant from each temperature. Cells were washed thrice with PBS and mounted in
well was carefully removed; formazan crystals were dissolved in medium containing DAPI. Images were captured using the
100 mL of DMSO and absorbance at 540 nm wavelengths was Olympus confocal microscope and analyzed with Provision
recorded. software.
S.P. Shaik et al. / Bioorganic & Medicinal Chemistry 25 (2017) 3285–3297 3297

4.2.6. Western blot analysis of soluble versus polymerized tubulin Supplementary data
Cells were seeded in 12-well plates at 1  105 cells per well in
complete growth medium. Following treatment of cells with Supplementary data associated with this article can be found, in
respective compounds (5f, 5k and nocodazole and paclitaxel) for the online version, at http://dx.doi.org/10.1016/j.bmc.2017.04.013.
a duration of 24 h, cells were washed with PBS and subsequently
soluble and insoluble tubulin fractions were collected. To collect References
the soluble tubulin fractions, cells were permeabilized with
200 mL of pre-warmed lysis buffer [80 mM Pipes-KOH (pH 6.8), 1. Amos LA. Org Biomol Chem. 2004;2:2153.
2. Mahindroo N, Liou JP, Chang JY, Hsieh HP. Expert Opin Ther Pat. 2006;16:647.
1 mM MgCl2, 1 mM EGTA, 0.2% Triton X-100, 10% glycerol, 0.1% 3. Downing KH, Nogales E. Curr Opin Cell Biol. 1998;10:16.
protease inhibitor cocktail (Sigma-Aldrich)] and incubated for 4. Hsieh HP, Liou JP, Lin YT, et al. Bioorg Med Chem Lett. 2003;13:101.
3 min at 30 °C. Lysis buffer was gently removed, and mixed with 5. Belotti D, Vergani V, Drudis T, et al. Clin Cancer Res. 1996;2:1843.
6. Mekhail TM, Markman M. Expert Opin Pharmacother. 2002;3:755.
100 mL of 3 Laemmli’s sample buffer (180 mM Tris-Cl pH 6.8, 7. Prota AE, Bargsten K, Northcote PT, et al. Angew Chem Int Ed. 2014;53:1621.
6% SDS, 15% glycerol, 7.5% b-mercaptoethanol and 0.01% bro- 8. Rowinsky EK, Donehower RC. Pharmacol Ther. 1991;52:35.
mophenol blue). Samples were immediately heated to 95 °C for 9. Nam NH. Curr Med Chem. 2003;10:1697.
10. Bhattacharyya B, Panda D, Gupta S, Banerjee M. Med Res Rev. 2008;28:155.
3 min. To collect the insoluble tubulin fraction, 300 lL of 1
11. Jordan MA. Curr Med Chem. 2002;2:1.
Laemmli’s sample buffer was added to the remaining cells in each 12. Porretta GC, Cerreto F, Fioravanti R, et al. Farmaco Sci. 1988;43:15.
well, and the samples were heated to 95 °C for 3 min. Equal vol- 13. Zuliani V, Fantini M, Nigam A, Stables JP, Patel MK, Rivara M. Bioorg Med Chem.
umes of samples were run on an SDS-10% polyacrylamide gel 2010;18:7957.
14. Fantini M, Rivara M, Zuliani V, et al. Bioorg Med Chem. 2009;17:3642.
and were transferred to a nitrocellulose membrane employing 15. a Tuyen TN, Sin KS, Kim HP, Park H. Arch Pharmacal Res. 2005;28:1013;
semidry transfer at 50 mA for 1 h. Blots were probed with mouse b Khanna IK, Yu Y, Huff RM, et al. J Med Chem. 2000;43:3168.
anti-human a-tubulin diluted 1:2000 ml (Sigma) and stained with 16. Plummer CW, Finke PE, Mills SG, et al. Bioorg Med Chem Lett. 2005;15:1441.
17. Bellina F, Cauteruccio S, Di Fiore A, Rossi R. Eur J Org Chem. 2008;32:5436.
rabbit anti-mouse secondary antibody coupled with horseradish 18. Bonezzi K, Taraboletti G, Borsotti P, Bellina F, Rossi R, Giavazzi R. J Med Chem.
peroxidase, diluted 1:5000 ml (Sigma). Bands were visualized 2009;52:7906.
using an enhanced Chemiluminescence protocol (Pierce) and 19. a Schobert R, Biersack B, Dietrich A, Effenberger K, Knauer S, Mueller T. J Med
Chem. 2010;53:6595;
radiographic film (Kodak). b Li WT, Hwang DR, Song JS, et al. J Med Chem. 2010;53:2409.
20. a Al-Tel TH, Al-Qawasmeh RA, Zaarour R. Eur J Med Chem. 2011;46:1874;
b Palkar M, Noolvi M, Sankangoud R, Maddi V, Gadad A, Nargund LV. Arch
4.2.7. Measurement of the mitochondrial membrane potential Pharm. 2010;343:353.
21. El-Shorbagi AN, Sakai S, El-Gendy MA, Omar N, Farag HH. Chem Pharm Bull.
The mitochondrial membrane potential was measured with 1989;37:2971.
lipophilic cationic dye JC-1 (Molecular Probes), as described.53 Cul- 22. Mase T, Arima H, Tomioka K, Yamada T, Murase K. J Med Chem. 1986;29:386.
tures were treated with the test drugs 5f and 5k at 0.5 mM for 48 h. 23. Ager IR, Barnes AC, Danswan GW, et al. J Med Chem. 1988;31:1098.
24. Srimanth K, Rao VR, Krishna DR. Arzneim-Forsch. 2002;52:388.
After drug treatment the MCF-7 cells were incubated with JC-1 dye
25. Trapani G, Franco M, Latrofa A, Reho A, Liso G. Eur J Pharm Sci. 2001;14:209.
for 20 min at 37 °C. After incubation cultures were used for mea- 26. Nobuaki A, Yukitaka I, Mayumi Y, et al. Cancer Lett. 2006;238:119.
suring the mitochondrial membrane potential (DWm),according 27. Chao Q, Sprankle KG, Grotzfeld KM, et al. J Med Chem. 2009;52:7808.
28. Kumbhare RM, Kumar KV, Janaki Ramaiah M, et al. Eur J Med Chem.
to the manufacturer’s instructions. The shift of the membrane
2011;46:4258.
potential was measured by flow cytometry using JC-1 (Molecular 29. Kamal A, Sultana F, Janaki Ramaiah M, et al. Chem Med Chem. 2012;7:292.
Probes), as previously described. 30. Suman P, Ramalinga Murthy T, Rajkumar K, et al. Eur J Med Chem. 2015;90:603.
31. Suman P, Dayakar C, Rajkumar K, et al. Bioorg Med Chem Lett. 2015;25:2390.
32. Raju G, Srinivas R, Damoder Reddy M, Raji Reddy Ch, Nagesh N. Nucleosides
Nucleotides Nucleic Acids. 2014;33:489.
4.2.8. Flow cytometric evaluation of apoptosis 33. Kamal A, Subba Rao AV, Vishnuvardhan MPS, et al. Org Biomol Chem.
MCF-7 were seeded in six-well plates and allowed to grow over- 2015;13:4879.
night. The medium was then replaced with complete medium con- 34. Thomas KD, Adhikari AV, Chowdhury IH, Sumesh E, Pal NK. Eur J Med Chem.
2011;46:2503.
taining 0.5 lM concentration of compounds, 5f and 5k for 48 h 35. He R, Chen Y, Chen Y, et al. J Med Chem. 2010;53:1347.
along with vehicle alone (0.001% DMSO) as the control. After 36. Giffin MJ, Heaslet H, Brik A, et al. J Med Chem. 2008;51:6263.
48 h of drug treatment, the cells from the supernatant and adher- 37. a Aher NG, Pore VS, Mishra NN, Kumar A, Shukla PK, Sharma A. Bioorg Med
Chem Lett. 2009;19:759;
ent monolayer cells were harvested by trypsinization, and washed b Patpi SR, Pulipati L, Yogeswari P, et al. J Med Chem. 2012;55:3911.
with PBS at 3000 rpm. Then the cells were stained with Annexin V- 38. Shaik SP, Nayak VL, Sultana F, et al. Eur J Med Chem. 2017;126:36.
FITC and propidium iodide using the Annexin-V-PI apoptosis 39. Abignente E, Arena F, De Caprariis P, Parente L. Farmaco Sci. 1977;32:735.
40. Alvarez SG, Alvarez MT. Synthesis. 1997;4:413.
detection kit (Sigma Aldrich). Flow cytometry was performed using 41. Botta M, Armaroli S, Castagnolo D, Fontana G, Perad P, Bombardelli E. Bioorg
a FACScan (Becton Dickinson) equipped with a single 488 nm Med Chem Lett. 2007;17:1579.
argon laser as described earlier.54 Annexin V-FITC was analyzed 42. Kanthou C, Greco O, Stratford A, et al. Am J Pathol. 2004;165:1401.
43. Maity HA, McKenna WG, Muschel RJ. J Biol Chem. 1995;270:28419.
using excitation and emission settings of 488 nm and 535 nm
44. Salaun P, Rannou Y, Prigent C. Adv Exp Med Biol. 2008;617:41.
(FL-1 channel); PI, 488 nm and 610 nm (FL-2 channel). 45. Rhind N, Russell P. Cold Spring Harbor Perspect Biol. 2012;4:005942.
46. Huber K, Patel P, Zhang L, et al. Mol Cancer Ther. 2008;7:143.
47. Kamal A, Subba Rao AV, Srinivas Reddy T, et al. MedChemComm. 2015;6:1842.
48. Gonda K, Tsuchiya H, Sakabe T, et al. Biochem Biophys Res Commun.
Acknowledgments
2008;370:629.
49. Zhu H, Zhang J, Hu NY, Yang B, He Q. Invest New Drugs. 2010;28:493.
S.P.S. and F.S., thank CSIR, New Delhi and the UGC, New Delhi 50. Ravelli RB, Gigant B, Curmi PA, et al. Nature. 2004;428:198.
for the award of fellowships. We acknowledge funding received 51. Morris GM, Huey R, Lindstrom W, et al. Comput Chem. 2009;30:2785.
52. Warren LD, The PyMOL Molecular Graphics System, DeLano Scientific LLC, San
from the project entitled ‘Affordable Cancer Therapeutics (ACT)’ Carlos, CA, http://www.pymol.org.
under XIIth five year plan and extend our appreciation to the Inter- 53. Romagnoli R, Baraldi PG, Cruz-Lopez O, et al. J Med Chem. 2010;53:4248.
national Scientific Partnership Program ISPP at King Saud Univer- 54. (a) Browne LJ, Eur Pat Appl, 1985; 165: 904, (Chem Abstr, 1986; 105: 6507d).;
(b) Browne LJ, Gude C, Rodriguez H, Steele RE, Bhatnager A. J Med Chem.
sity for funding this research work through ISPP#0054. 1991;34:725.

You might also like