You are on page 1of 19

Arabian Journal of Chemistry (2019) 12, 5000–5018

King Saud University

Arabian Journal of Chemistry


www.ksu.edu.sa
www.sciencedirect.com

ORIGINAL ARTICLE

Pharmacophore modeling, 3D-QSAR,


docking study and ADME prediction of acyl
1,3,4-thiadiazole amides and sulfonamides as
antitubulin agents
Mohemmed Faraz Khan a, Garima Verma a, Wasim Akhtar a,
Mohammad Shaquiquzzaman a, Mymoona Akhter a, Moshahid Alam Rizvi b,
Mohammad Mumtaz Alam a,*

a
Drug Design and Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard,
New Delhi 110062, India
b
Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India

Received 3 September 2016; accepted 8 November 2016


Available online 16 November 2016

KEYWORDS Abstract Pharmacophore modeling, molecular docking, and in silico ADME studies have been car-
3D-QSAR; ried out to determine the binding mode and drug likeliness profile of acyl 1,3,4-thiadiazole amides and
Pharmacophore modeling; sulfonamides as antitubulin agents. A four point pharmacophore model (AAHR.11) was generated
Molecular docking; using 63 compounds with IC50 values ranging from 3.16 to 505.76 lM. A statistically significant
In silico ADME; 3D-QSAR model was generated from the pharmacophore hypothesis. The model had a high correla-
Tubulin tion coefficient (R2 = 0.8925), cross validation coefficient (Q2 = 0.8204) and F value (44.3) at 6 com-
ponent PLS factor. The results of external validation indicated that the generated QSAR model
possessed a high predictive power (R2 = 0.83). The generated model also passed Tropsha’s test for pre-
dictive ability and Y-Randomisation test. The Domain of Applicability (APD) of the model was also
successfully defined to ascertain that a given estimation can be considered reliable. Further, the restric-
tivity of the model was checked with inactive compounds by enrichment studies using the decoy test. In
order to evaluate the effectiveness of the docking protocol, co-crystallized ligand was extracted from
the ligand binding domain of the protein and was re-docked into the same position. The conformer
obtained on re-docking and the co-crystallized ligand were superimposed and the RMSD between

Abbreviations: R2, regression coefficient; Q2, Cross Validation Correlation Coefficient; Gscore, Glide score; RMSD, root mean square deviation; A,
hydrogen bond acceptor; R, aromatic ring; H, hydrophobic; PLS, Partial Least Square; APD, Domain of Applicability; SD, standard deviation.
* Corresponding author. Fax: +91 11 26059663.
E-mail address: drmmalam@gmail.com (M.M. Alam).
Peer review under responsibility of King Saud University.

Production and hosting by Elsevier

http://dx.doi.org/10.1016/j.arabjc.2016.11.004
1878-5352 Ó 2016 The Authors. Production and hosting by Elsevier B.V. on behalf of King Saud University.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5001

the two was found to be 0.853 Å. ADME predictions were also performed for these compounds. Out-
comes of the present study have been first utilized to get insight into the molecular feature that pro-
motes bioactivity, and then within screening procedure, have been exploited for the estimation of
novel potential antitubulin compounds prior to their synthesis and biological tests.
Ó 2016 The Authors. Production and hosting by Elsevier B.V. on behalf of King Saud University. This is
an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction 2. Materials and methods

Cancer continues to be the cause of a quarter of deaths in developed 2.1. Data set
countries. At present, in United States, it is the second leading reason
for deaths and in near future, it is expected to surpass heart diseases
and become the most leading cause of deaths (Shobeiri et al., 2016). A set comprising of 63 compounds bearing cinnamic acyl
With the urgent need for development of novel and effective anticancer 1,3,4-thiadiazole amides and sulfonamides was taken from
agents, numerous targets are being explored by scientists. the available literatures (Yang et al., 2012; Luo et al., 2011)
Microtubules, formed by polymerization of tubulin heterodimers and was used in the present study. The selected compounds
are the key constituents of the cytoskeleton of eukaryotic cells. They for the data set shared the same assay procedure (Hamel,
play a vital role in diverse cellular functions such as mitosis, exocytosis 2003; Hamel and Lin, 1984) with significant variations in their
and maintenance of cellular morphology, active transport, cell shape structures and potency profiles. Inhibitory potencies of the
and polarization (Kamal et al., 2016; Zghaib et al., 2016). Thereby,
compounds included in data set, had IC50 values varying from
tubulin is considered as an imperative target for anticancer drug dis-
3.16 to 505.76 lM which were converted into molar values.
covery. Various antimitotic agents that interfere with normal dynamics
of tubulin polymerization and depolymerization include taxanes, vin- These were then converted into pIC50 values using the formula
blastine and colchicines (Abbas et al., 2016; An et al., 2016). given below.
Microtubules comprise of a/b tubulin heterodimers, which are in a
state of equilibrium (Kamal et al., 2014; Jordan et al., 1998). Micro-
pIC50 ¼ log10 ½IC50 
tubule targeting agents, interfering with microtubule dynamic stability, The 3D structures of ligands were generated using the
are widely employed for treatment of variety of cancers (Mollinedo builder panel in Maestro and subsequently optimized using
and Gajate, 2003; Zhou and Giannakakou, 2005). These agents are
LigPrep module (v3.1, Schrödinger 2016-1). Partial atomic
also known to arrest cell division during interphase.
charges were ascribed and possible ionization states were gen-
a and b tubulin share identical structures. Density maps for both
the subunits are almost superimposable with some differences limited erated at a pH of 7.0. The OPLS_2005 force field was used for
to the length and conformation of some loops, very slight displace- optimization for production of low energy conformer of the
ments (1 Å) of some of the secondary structure elements, and differ- ligand (Shivakumar et al., 2010). The energy minimization
ences in side-chain densities. Core of each monomer is comprised of was performed for each ligand till it reached a root mean
two b-sheets which are surrounded by a helices. The structure of each square deviation (RMSD) cutoff of 0.01 Å. The resulting
monomer can be divided into three functional domains: (i) the amino- structures were then taken for performing modeling studies.
terminal domain containing the nucleotide-binding region, (ii) an inter-
mediate domain containing the Taxol-binding site and (iii) the 2.2. Pharmacophore 3D-QSAR modeling
carboxy-terminal domain, which probably is the part of binding sur-
face for motor proteins. However, the main binding site of drugs is
the b-subunit, residues C356 and C241 and the region 1–36. Colchicine Phase (v4.6, Schrödinger 2016-1) was used for generation of
binds close to the interface of ab dimer (Nogales et al., 1998; Uppuluri pharmacophore and 3D-QSAR models for antitubulin agents
et al., 1993; Shearwin and Timasheff, 1994). (Dixon et al., 2006). The prepared ligands were imported for
A number of antitubilin agents such as Vinblastine, Vincristine, developing pharmacophore model panel of Phase with their
Vinorelbine, Vinflunine, Colchicine, Combretastatin, Paclitaxel, Doc- respective biological activity values. The ligands were assigned
etaxel and Epothilone find clinical application in the treatment of as actives with a threshold of pIC50 > 5.1 and inactives with a
numerous cancers (Mahindroo et al., 2006; Negi et al., 2015;
threshold of pIC50 < 3.45. Remaining compounds were con-
Dumontet, 2011). However, resistance to these agents and associated
sidered as moderately active. Phase (v4.0) has been proven to
side effects limit the use of these agents (Dumontet and Sikic, 1999;
Dumontet and Jordan, 2010). Presently, based on ligand and structure be an important tool for flexible ligand superposition (Sastry
based approaches, computational studies are considered effective tools et al., 2011; Miller et al., 1999). In the current study, we used
in medicinal chemistry, useful to accelerate the drug design process phase (v4.0) shape screening for flexible alignment of the
(Cichero et al., 2014, 2016, 2011a,b; Cichero and Fossa, 2012). Previ- selected antitubulin compounds. The most active compound
ously also researchers have reported computational work on anti- 25 was taken as the template and pharmacophore type volume
tubulin agents (Marzago and Chilin, 2014; Liao et al., 2010; Li scoring was kept as default. The default settings were used with
et al., 2015; Lokwani et al., 2013; Kandakatla et al., 2014). With these a maximum of 10 conformations per rotatable bonds and with
facts, we herein report pharmacophore modeling, 3D-QSAR, molecu- this a maximum of 100 conformers were generated. Conforma-
lar docking and ADME prediction of cinnamic acyl 1,3,4-thiadiazole
tions of these compounds were varied and at most one align-
amides and sulfonamides to provide an insight into the key structural
ment for every ligand was retained. For assigning training
features required for designing potential tubulin polymerization inhibi-
tors. The model developed in this study is a quite reliable one for all and test set for 63 compounds, random selection was made
predictions as it has been validated using different techniques. On using the Automated Random Selection option present in
the basis of the developed model, various novel active compounds tar- Phase (v4.0) software. Test set comprised of 18 compounds
geting tubulin have been identified via virtual screening. and remaining 45 compounds were included in training set.
5002 M.F. Khan et al.

Pharmacophore sites for these compounds included default set


Table 1 Intersite distances between the pharmacophoric sites
of chemical features of Phase: two hydrogen bond acceptors
of AAHR.11.
(A), aromatic ring (R) and a hydrophobe (H). Pharmacophore
matching tolerance was set as 1 Å. A total of 6 variants were Entry Site 1 Site 2 Distance
generated by keeping 5 and 4 as the maximum and minimum AAHR.11 A1 A5 5.074
number of sites respectively. It was specified that at least 5 AAHR.11 A1 H8 12.571
of the actives must match. Finally, it resulted in 10 possible AAHR.11 A1 R11 9.703
combinations of features which could give rise to common AAHR.11 A5 H8 8.957
pharmacophores. The resulting hypotheses were scored and AAHR.11 A5 R11 5.754
ranked in accordance with their vector, volume, site scores, AAHR.11 H8 R11 3.216
survival scores and survival actives (Dixon et al., 2006) shown
in Table 3.
generated common pharmacophore is shown in Fig. 2a and b.
In the generated hypotheses, four sites were found to be
Using the model, AAHR.11, fitness score of all the ligands was
common for all selected compounds. 3D-QSAR model was
evaluated (Table 5). Contour plot analysis was also done to
generated using Partial Least Square (PLS) regression statistics
identify the distinct imperative pharmacophoric requirements
and keeping the grid spacing of 1 Å. The number of PLS fac-
at spatial sites of structure.
tors included in model development is 6 because an incremen-
tal increase in the statistical significance and predictivity was
2.3. Model validation
observed till 6 PLS factor (Table 4).
The distance and angles between different sites of the model
AAHR.11 are shown in Fig. 1a and 1b. This is also presented The QSAR model AAHR.11 with 6 component PLS factor
in Tables 1 and 2. Alignment of actives and inactives with the was characterized as the best model. Pharmacophoric model

Figure 1 Intersite (a) distances and (b) angles between the pharmacophoric points of model AAHR.11. All distances are in Å unit. Pink
spheres with arrow, hydrogen bond acceptor (A); orange open circle, aromatic ring (R); green sphere, hydrophobe.
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5003

The predictive power of the produced 3D-QSAR model


Table 2 Intersite angles between the pharmacophoric sites of
was also assessed by using Enalos Model Acceptability Criteria
AAHR.11.
KNIME node (Melagraki and Afantitis, 2013), which includes
Entry Site 1 Site 2 Site 3 Angle all proposed tests by Tropsha (Zhang et al., 2006). Criteria fol-
AAHR.11 A5 A1 H8 35.7 lowed in developing activity/property predictors, especially for
AAHR.11 A5 A1 R11 28.2 continuous QSAR, are as follows: (i) correlation coefficient R
AAHR.11 H8 A1 R11 7.5 between the predicted and observed activities; (ii) coefficients
AAHR.11 A1 A5 H8 124.9 of determination (predicted versus observed activities R20 and
AAHR.11 A1 A5 R11 127.2 observed versus predicted activities R0 20 for regressions through
AAHR.11 H8 A5 R11 2.3 the origin); and (iii) slopes k and k0 of regression lines through
AAHR.11 A1 H8 A5 19.3
the origin. The criteria for the acceptability of the QSAR
AAHR.11 A1 H8 R11 23.4
AAHR.11 A5 H8 R11 4.0
model are represented in left column of Fig. 5.
AAHR.11 A1 R11 A5 24.6 Further validation of the model was done by enrichment
AAHR.11 A1 R11 H8 149.1 studies using the decoy test. 1000 decoy test set compounds
AAHR.11 A5 R11 H8 173.7 (Kirchmair et al., 2008) retrieved from the Schrödinger data
base were taken to evaluate predictive power of the model.
Enrichment factor (EF) and Robust initial enhancement
(RIE) were calculated to ensure the reliability of the model
was validated by its accuracy in estimation of the activity of and for the accurate ranking of compounds (Sheridan et al.,
training set ligands. Scatter plots for experimental and pre- 2001).
dicted activities of ligands elicited significant linear correlation
and moderate difference between the experimental and pre- 2.4. Domain of applicability
dicted values shown in Fig. 3a and b. The efficacy of model
AAHR.11 was also examined with external validation. Biolog- In order to screen a QSAR model for new compounds, its
ical test used for compounds of both the ‘‘internal” training domain of application (APD) (Tropsha et al., 2003; Shen
and test sets was the same as used for compounds of the ‘‘ex- et al., 2004) must be defined and predictions for only those
ternal test set”. In order to clearly distinguish between the test compounds that fall into this domain may be considered reli-
set and external test set, it is mandatory to mention that com- able. Extent of Extrapolation (Tropsha et al., 2003) is one of
pounds of training and test share structural similarity while the simple approaches based on the calculation of the leverage
compounds of external test set are from a different publica- (hi) for each chemical, where the QSPR model is used to pre-
tion. This external validation was done to check further relia- dict its activity.
bility of the model (Table 1: Supplementary Material and
1
Fig. 4). Graphs of actual value vs. predicted value and residual hi ¼ xi XT X xTi
value vs. predicted value were plotted.

Table 3 Score of different parameters of the hypotheses.


S. no. Hypothesis Survival score Survival inactive Site Vector Matches Activity Inactive
1. AAHR.11 3.640 1.921 0.87 0.966 5 5.500 1.719
2. AAHR.7 3.621 1.865 0.86 0.967 5 5.137 1.755
3. AAHR.9 3.597 1.805 0.85 0.967 5 5.107 1.792
4. AAHR.8 3.581 2.513 0.82 0.954 5 5.500 1.068
5. AAHR.10 3.544 1.956 0.79 0.793 5 5.137 1.588
6. AAHR.12 3.520 2.365 0.78 0.956 5 5.107 1.156
A: Acceptor; H: Hydrophobic; R: Aromatic ring.

Table 4 PLS statistical parameters of the model AAHR.11.


PLS SD R2 F P Stability RMSE Q2 Pearson-R
1 0.4436 0.4627 31.9 1.899e006 0.9890 0.3794 0.3287 0.6088
2 0.3599 0.6560 34.3 4.557e009 0.6025 0.311 0.5489 0.814
3 0.2916 0.7805 41.5 1.286e011 0.7927 0.2854 0.6202 0.857
4 0.2669 0.8213 39.1 2.904e012 0.7914 0.2489 0.7111 0.8866
5 0.2437 0.8555 39.1 6.229e013 0.7491 0.2057 0.8026 0.9168
6 0.2134 0.8925 44.3 3.959e014 0.7355 0.1962 0.8204 0.9124
SD: Standard deviation of regression, R2: Regression coefficient; F: Ratio of the model variance to the observed activity variance (variance
ratio); P: Significance level of variance ratio; Q2: Cross validated correlation coefficient for the test set; RMSE: the RMS error in the test set
predictions.
5004 M.F. Khan et al.

Figure 2 (a) Mapping of the active compounds onto the pharmacophore. (b) Mapping of inactive compounds onto the pharmacophore.

In the above equation, xi is the row vector containing the k 2.5. Y-Randomization test
model parameters of the query compound and X is the n  k
matrix containing the k model parameters for each one of Y-Randomization technique ensures the robustness of a QSPR
the n training compounds. A leverage value greater than model (Tropsha et al., 2003; Shen et al., 2004). The dependent
3k/n is considered large. It means that the predicted response variable vector is randomly shuffled and a new QSPR model is
is the result of a substantial extrapolation of the model and generated. The procedure is repeated several times and the new
may not be reliable. QSPR models are expected to have low R2 and Q2 values. In
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5005

Table 5 Calculated pIC50 for compounds in predicted set.


Ligand Chemical structure Experimental activity (pIC50) Predicted activity (pIC50) Residual activity Fitness
O N N

1t N S 4.75 4.89 0.14 2.37


H

O N N

2 N
H
S 4.74 4.81 0.07 2.66
F
F F

O
3 4.83 4.77 0.06 2.73
HN N
N
S

O N N

4t
N S 4.85 4.88 0.03 2.73
H

N N O

t S N-
5 5.02 4.50 0.52 1.86
O

N N O

6 Cl S N 4.71 4.78 0.07 2.40


H

N N O

7 Cl S N
H 4.70 4.75 0.05 2.69
F
N N O
Cl
S N
8 H F 4.71 4.66 0.05 2.75
F
F
N N O

9 Cl S N 4.87 4.83 0.04 2.76


H

N N O

Cl S N
10 H 4.81 5.02 0.21 2.95
O

N N O

11t Br
S N 4.73 4.80 0.07 2.40
H

N N O
Br
12 S N
H
4.66 4.71 0.05 2.69
F
N N O
Br
S N
13 H
F
4.63 4.66 0.03 2.75
F F
N N O
Br
14t S N 4.90 4.79 0.11 2.74
H

N N O
Br
S N
15 H 5.00 5.04 0.04 2.95
O

N N O

16 S N 4.82 4.88 0.06 2.40


H

N N O

17t S N
H
4.82 4.86 0.04 2.69
F
N N O

S N
18t H
F
4.87 4.76 0.11 2.75
F F

(continued on next page)


5006 M.F. Khan et al.

Table 5 (continued)
Ligand Chemical structure Experimental activity (pIC50) Predicted activity (pIC50) Residual activity Fitness
N N O

19 S N 4.90 4.92 0.02 2.74


H

N N O

S N
20 H 5.12 5.11 0.01 2.95
O

N N O

21 O S N 5.04 5.12 0.08 2.45


H

N N O
O
22 S N
H
5.06 5.08 0.02 2.72
F
F F

23 HN N 5.10 5.04 0.06 2.76


N
S

O
N N O

24 O S N 5.13 5.15 0.02 2.77


H

N N O
O
S N
25 H 5.50 5.15 0.35 3.00
O

O O
26 S 3.90 3.86 0.04 1.64
N-
O

F
O O
t
27 S
N-
4.83 4.79 0.04 1.64
O

Cl
O O
28 S
N-
4.82 4.54 0.28 1.64
O

Br
O O
29 S
N-
3.73 4.36 0.63 1.64
O

O S O
N- O

30 4.83 4.78 0.05 1.86

O S O
O N-
31 3.90 4.19 0.29 1.84

F
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5007

Table 5 (continued)
Ligand Chemical structure Experimental activity (pIC50) Predicted activity (pIC50) Residual activity Fitness
F
O O
32 S 5.12 5.11 0.01 1.84
N-
O
F
Cl
O O
33 S 4.87 4.81 0.06 1.85
N-
O
F
Br
O O
34t N-
S 4.41 4.59 0.18 1.85
O
F

O S O
N- O

35 4.32 4.30 0.02 1.92

Cl

O S O
O N-
36 4.04 3.73 0.31 1.91

Cl
F

O S O
O N-
37 4.58 4.65 0.07 1.91

Cl
Cl
O O
t S
38 N- 4.58 4.33 0.25 1.91
O
Cl
Br
O O
t S
39 N- 4.56 4.19 0.37 1.91
O
Cl

O S O
N- O

40 3.69 3.92 0.23 1.94

Br

O S O
O N-
41 3.44 3.45 0.01 1.93

Br

(continued on next page)


5008 M.F. Khan et al.

Table 5 (continued)
Ligand Chemical structure Experimental activity (pIC50) Predicted activity (pIC50) Residual activity Fitness
F

O S O
O N-
42 5.00 4.44 0.56 1.92

Br
Cl

O S O
O N-
43 3.56 4.12 0.56 1.93

Br
Br
O O
44t N-
S 3.92 3.98 0.06 1.93
O
Br

O S O
N- O

45 3.29 3.66 0.37 1.89

O O
S
46 N- 3.35 3.25 0.10 1.87
O
O
F
O O
47t N-
S 3.87 4.04 0.17 1.87
O
O
Cl
O O
48t N-
S 3.82 3.79 0.03 1.88
O
O
Br
O O
49 S 3.87 3.68 0.19 1.88
N-
O
O

O S O
N- O

50 4.10 4.21 0.11 1.88

O O
S
51 N- 3.96 3.73 0.23 1.86
O
N

F
O O
S
52 N-
O 4.50 4.56 0.06 1.86
N
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5009

Table 5 (continued)
Ligand Chemical structure Experimental activity (pIC50) Predicted activity (pIC50) Residual activity Fitness
Cl
O O
S
53t N-
O 4.08 3.98 0.10 1.87
N

-
O O
N+

54t 3.82 3.70 0.12 1.82


N O-
O S O

O
-O
N+
O
55 N
S
3.35 3.52 0.17 1.67
O - O

O
- N+
O
O
56 N 4.74 4.56 0.18 1.65
S
O - O
F
O
- N+
O
O
57 N
S 3.43 3.49 0.06 2.12
O - O
Cl
O
-
N+
O
O
58 N 3.40 3.39 0.01 0.72
S
O- O
Br

O S O
-
O N
59 3.65 3.83 0.18 1.94
O
- N+
O

O O-
O O- N+
60 S
N
3.53 4.02 0.49 1.04
O

F
O O-
O O- N+
t
61 S
N
3.70 3.34 0.36 1.57
O

Cl
O O-
O O- N+
62 S
N
3.67 3.53 0.14 1.87
O

Br
O O-
O O- N+
63 t
S
N
3.48 3.57 0.09 0.72
O

t
: test.

case of opposite results, an acceptable QSPR model cannot be range of targets. We decided to use PubChem database within
obtained. our in silico framework for virtual screening compounds which
could act as tubulin inhibitors. For this purpose, we developed
2.6. Virtual screening a KNIME workflow to assess the compounds that will be pri-
oritized for virtual screening. Our first aim was to mine all
PubChem database serves as an important source of com- compounds having structural similarity with our set of com-
pounds which could be prioritized for screening for a wide pounds within PubChem database. To achieve that, we have
5010 M.F. Khan et al.

Figure 3 (a): Scatter plot of the observed versus phase-predicted activity for (a) training set and (b) test set compounds with best fit line
y = 0.84x + 0.66 (R2 = 0.83).

used Substructure Matcher node included in Indigo chemin- box was generated at a centroid of active site. For docking,
formatics toolkit. All these compounds were compared to the low energy conformations of all the compounds were docked
most active compound of our original data set in terms of sim- into the catalytic domain of tubulin protein (PDB Code:
ilarity given by Indigo Fingerprints. It is known that similarity 1SA0) using Grid based Ligand Docking with Energetics
searching refers to the calculation of the similarity coefficients (Glide v7.0, Schrödinger 2016-1) (Friesner et al., 2006) in extra
for a given molecule and each compound included within a precision mode without applying any constraint. The best
database. Among the different proposed methodologies, simi- docked structure was identified using Glide score function,
larity searching using fingerprints is one of the most widely Glide energy and Glide Emodel energy (Table 8: Supplemen-
applied methods for ligand-based screening. Fingerprints are tary Material). The lowest energy docked compound 25 was
representations of the structural features of a molecule given selected for further studies. A map of hydrophobic and hydro-
as a series of binary digits (bits) that account for the presence philic fields for inhibitor 25 was also generated (Fig. 8).
or absence of particular substructures in the molecule. For this
study, Fingerprint Similarity node using Tanimoto metric for 2.8. Lipinski’s rule for drug likeliness and in silico ADME
similarity computation was used to afford the most similar prediction
compounds that could be prioritized for screening.
All the retrieved compounds were virtually screened with
We further predicted the drug-like behavior of the compounds
the ultimate goal of prioritizing the most promising new syn-
through the analysis of pharmacokinetic profile of the com-
thetic targets. The proposed 3D-QSAR model was used to
pounds by using Qikprop module (v4.3, Schrödinger 2016-1).
identify the most active compounds that are proposed for fur-
The compounds prepared by LigPrep module (v3.1, Schrödin-
ther investigation. For this, find matches option of Phase mod-
ger 2016-1) were utilized for the calculation of pharmacoki-
ule was used.
netic parameters by QikProp v4.3. Physically significant
descriptors and pharmaceutically relevant properties of all
2.7. Molecular docking
the test compounds such as molecular weight, log P, H-bond
donors, and H-bond acceptors were analyzed in accordance
The catalytic domain of tubulin enzyme in complex with col- with Lipinski’s rule of five (Lipinski et al., 2001).
chicine (PDB Code: 1SA0) was obtained from protein data
bank and prepared using the protein preparation wizard 3. Results and discussion
(Sastry et al., 2013) available in Schrödinger suite 2016-1.
Crystallographic water molecules i.e. without 3H bonds were
3.1. Pharmacophore and 3D-QSAR models
deleted and hydrogen bonds corresponding to pH 7 were
added considering the appropriate ionization states for both
acidic and basic amino acid residues. OPLS_2005 force field Phase (v4.0) (Dixon et al., 2006) module of Schrödinger 2016-1
was used for energy minimization of the crystal structure was used for the development of pharmacophore model. Atom
(Shivakumar et al., 2010). Active site was defined with a radius based 3D-QSAR revealed the effect of substituents on activity.
of 14 Å around the ligand present in crystal structure. Grid Based on the sites, a maximum of four features were allowed
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5011

Figure 4 (a) Plot of actual value vs. predicted value of external training set y = 0.84x + 0.66 (R2 = 0.83). (b) Plot of residual value vs
predicted value.

3.2. Model validation

The predictability and validity of the common pharmacophore


model, AAHR.11 (test set), based on active compounds were
judged by cross validation coefficient (Q2 = 0.8204) (Table 4).
Regression coefficient (R2) of the training set was 0.89, which
exhibited relevance of the model. Stability of the generated
model ranges from 0.7355 to 0.9890 on the maximum scale
of 1. The F value was found to be 44.3. Additionally, P value
of 3.959e-014 and Pearson r of 0.9124 indicated greater degree
of confidence on the model. Standard deviation (SD) value of
0.2134 and root mean square error (RMSE) of 0.1962 indicate
the stability of the generated model for estimation of unknown
compounds in the test. In order to evaluate the efficacy of the
generated model, it was further validated using an external test
set (Li et al., 2014). The calculated pIC50 values of the com-
pounds included in predicted set are given in Table 5. Predicted
Figure 5 Screenshot of results obtained by (a) Enalos Model
values for external test set are given in Table 1 (Supplementary
Acceptability Criteria KNIME node; (b) Enalos Domain-Lever-
Material). A plot of experimental vs. predicted pIC50 of exter-
age node.
nal test set is shown in Fig. 4a and a plot of residual vs. pre-
dicted value is shown in Fig. 4b. These two plots are vital
for developing the hypotheses. For this, 6 common hypotheses for the predictive ability of QSAR. Scatter residual plots were
were generated in all for 63 compounds (Table 3). The best fit- used to identify the outliers from the QSAR model (Golbraikh
ted model AAHR.11 (R2 = 0.8925, Q2 = 0.8204 and and Tropsha, 2002a,b). Fig. 4b elicits that there is no outlier in
F = 44.3) consists of two hydrogen bond acceptor, one the study. The presence or absence of outliers can be easily
hydrophobic and aromatic ring features (Fig. 1a and b) with determined from the scatter plot by comparing the number
highest survival score (3.640). This is also apparent from the of compounds taken in the test set and number of dots present
comparison made by the deduction of survival inactive from in the plot. In this case, both the numbers were same, thereby
survival active. It deducts inactive features from the hypothesis rendering the model stable and reliable. It was able to support
and was decisively highest for the hypothesis AAHR.11. the experimental pIC50 values for compounds in the external
Among the pharmacophore features, intersite distances and test set. Predictive correlation coefficient R2 value of 0.83
angles between site points were found to be the principal attri- was observed for the external test set for the developed QSAR
bute and the point of difference between active and inactive is model. R2 value of more than 0.5 between the predicted and
due to the interstitial site distances as evident by the pharma- experimental values renders the model to be good and was able
cophore hypothesis AAHR.11 alignment over active to predict the inhibitory activity of compounds not included in
(pIC50 > 5.1) and inactive compounds (pIC50 < 3.45) in the model development process (Golbraikh and Tropsha,
Fig. 2a and b respectively. 2002a,b).
5012 M.F. Khan et al.

Figure 6 QSAR model visualized in the context of favorable and unfavorable hydrogen bond donor effects in (a) compound 25 (5.50)
and (b) compound 45 (3.29). QSAR model visualized in the context of favorable and unfavorable hydrophobic interactions in (c)
compound 25 (5.50) and (d) compound 45 (3.29). QSAR model visualized in the context of favorable and unfavorable electron
withdrawing groups in (e) compound 25 (5.50) and (f) compound 45 (3.29).

Furthermore, the Enalos Model Acceptability Criteria the Schrödinger database. The model was able to find 100%
KNIME node has also been applied to the data. The model of active compounds in the hit list. RIE was calculated for
passed Tropsha’s recommended tests for predictive ability the generated models to estimate the contribution of the active
and the results are depicted in Fig. 5a. These results suggest compounds ranking in the enrichment. For AAHR.11, RIE
that this alignment can effectively take into consideration the value of 8.24 indicated the superiority of the pharmacophore
ligand-receptor interactions, and the QSAR model is thus reli- model ranking over random distribution. AUC of the ROC
able and could be used in the design of new tubulin inhibitors curve is also considered as a reliable factor to assess the perfor-
within this structural motif of molecules. mance of the developed model. AAHR.11 showed a good
In order to validate the discriminatory ability, the model value of 0.95 AUC and 0.99 ROC (Fig. 1 and Tables 2–5 of
was screened against 1000 decoy molecules retrieved from Supplementary Data).
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5013

Figure 7 (a) Binding mode of compound 25 in the catalytic pocket of 1SA0, (b) 2D-ligand interaction diagram of 25 in the catalytic
pocket of 1SA0, (c) Hydrophobic interactions of 25 in the catalytic pocket of 1SA0, (d) Overlay of docked pose (magenta) of colchicine
with its crystal structure conformation (pink).

3.3. Domain of applicability After model validation, the APD of our model was also
defined to ascertain that a given estimation can be considered
It is important that the limitations of the model are also reliable. Enalos Domains – Leverage (Extent of Extrapolation)
described via the APD. This gives an important indication as node that executes the aforementioned procedure is included in
the user can freely and creatively design novel molecules but our workflow and was used to assess APD of the proposed
will be warned for the reliability of the estimation when the model. In this method, the APD limit was defined as 0.167.
structural characteristics cannot be tolerated by the model. This limit was defined on the basis of equation provided in
5014 M.F. Khan et al.

Figure 8 Map of hydrophobic and hydrophilic fields for inhibitor 25 into the catalytic protein of tubulin (1SA0).

Methods section. In the APD, all the compounds fell within concordance with those reported by the researchers who had
the range i.e. none of the compounds was in the group of out- synthesized and biologically evaluated these compounds
liers (Fig. 5b). All these compounds were in the domain of the (Yang et al., 2012; Luo et al., 2011).
applicability, and hence can be considered reliable. The hydrogen-bond donor nature for the most active com-
pound 25 (Fig. 6a) and the least active compound 45 (Fig. 6b)
3.4. Y-Randomization test when compared showed their most favorable region with blue
color and unfavorable regions with red color. Hydrogen-bond
donor mapping revealed that favorable regions lied near the
The model was further validated by applying Y-randomization.
nitrogen atoms of thiadiazole, indicating their importance
Ten random shuffles of the Y vector were performed and the
for activity as compared to the least active compound 45 bear-
low R2 and Q2 values were obtained. This showed that the good
ing sulfone group. Therefore, the presence of thiadiazole ring
results in the original model were not due to a chance correla-
with hydrogen donor group in the scaffold backbone is vital
tion or structural dependency of the training set. The R2 and Q2
for the tubulin inhibitory activity. Almost all the thiadiazole
values were in the range of 0.05 to 0.345 and 0.00 to 0.11 respec-
containing compounds were found to have better activity pro-
tively. It should be noted that for each random permutation of
file in comparison with the ones with sulfone moiety.
the Y vector, the complete training procedure was followed for
Another significant component that impacts the antitubulin
developing the new QSPR model, including the selection of the
activity is the hydrophobic character. Fig. 6c and d when com-
most appropriate descriptors.
pared for their hydrophobic nature for the most active com-
pound 25 and least active compound 45 reveals that the blue
3.5. 3D-QSAR contour map analysis region, an indicative of hydrophobic character, around the thia-
diazole ring is essential for a compound to be an antitubulin
Contour plot analysis was performed to identify the effect of agent. In Fig. 6d, the presence of red cubes at para position of
spatial arrangement of structural features such as electrostatic, phenyl ring directly attached to the sulfone group indicates that
ionic, hydrophobic, H-bond donor and H-bond acceptor hydrophobic groups are unfavourable at this position. This
regions on antitubulin activity. Their individual positive con- assumption is supported by the low activity of methyl substi-
tribution is shown in blue cubes and negative contribution is tuted compounds when compared to their unsubstituted deriva-
indicated by red cubes. Fig. 6a–f shows the comparison of tives. This is evident while comparing the compounds 31 with 30,
most significant favorable and unfavorable interactions, which 36 with 35, 41 with 40, 51 with 50, 55 with 54 and 60 with 59.
arise on the application of QSAR model to the most active and In contour plot of compound 25 (Fig. 6e), the presence of
least active ligands. Results of our study were found to be in red cubes at para position of phenyl ring attached directly to
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5015

Figure 9 Superimposition of conformations of inhibitor 25: best docking pose and pose of the AAHR.11 model (RMSD: 0.870 Å).

the thiadiazole moiety indicates that the presence of electron Tanimoto similarity metric of over 0.80 were selected to assess
withdrawing groups is undesirable at this position. This is sup- the potency of these compounds. Our approach that is based
ported by less activity of compound 6 (pIC50 4.71) having Cl on the scaffold and fingerprint similarity search within Pub-
group at para position. A comparison of the para- Chem database resulted in a narrower chemical space that
substituents on the same ring depicted that an electron- increases the chance of success.
donating group (16, 21) (pIC50 4.82, 5.04) have slightly The pIC50 values of the 13 most potent compounds (Vs1–
improved antitubulin activity with the potency order Vs13) predicted with the established pharmacophore model
OCH3 > CH3. On the contrary, the presence of blue cubes are listed in Table 6 of Supplementary Material. All the virtual
at para position of cinnamoyl moiety indicated the preference screening compounds were within the domain of the applica-
of electron withdrawing groups at this position. This is bility of the model, and therefore their activity estimations
explained by the significant antitubulin activity of compounds can be accepted with confidence. The predicted compounds
with para halogen substitution (30, 35, 40) (pIC50 4.83, 4.32, were searched for their activity using the PubChem database
3.69) in the order of F > Cl > Br. The results demonstrated and their biological data have been reported in Table 6 (Sup-
that an electron-withdrawing group may have slightly plementary Material).
improved antitubulin activity. From the values themselves, it On the whole, obtained hits can serve as new chemical start-
is observed that the compounds substituted with electron ing points for further structural optimization of tubulin
donating groups (compound 45: pIC50 3.29) had relatively inhibitors.
lower pIC50 values in comparison with the compounds with
electron withdrawing substituents and to the unsubstituted 3.7. Molecular docking
compound 26 (pIC50 3.90).
The scores of docking studies are shown in Table 7 (Supple-
3.6. Virtual screening mentary Material). From 63 listed total-scores, pIC50 of most
compounds were in accordance with the Glide score. Docking
As described in the Materials and Methods section, the pro- study revealed that interactions were dominated by the
duced QSAR model can be used as a useful tool for screening hydrophobicity and aromaticity due to the presence of cin-
existing databases or virtual libraries in an effort to prioritize namoyl moiety. The interactions were dominated in the region
potent compounds for experimental evaluation. In this con- of Tyr 202, Ala 316, Ala 250 and Val 239 amino acid residues
text, a virtual screening of structurally similar compounds content due to pronouncing existence of active site in the
included in PubChem database was carried out using a region (Fig. 7a and b). In general, phenyl ring directly attached
KNIME workflow. to thiadiazole ring exhibited hydrophobic interactions with
First, 53,691 compounds were retrieved from the PubChem Lys 254, Leu 242, Ala 250, Leu 248, Ala 354, Val 318, Lys
database. Lipinski’s rule of five was used to scrutinize these 352, Ile 378 and Leu 255, the key residues necessary for bind-
compounds. A total of 47,615 compounds were obtained after ing of inhibitors (Fig. 7c). This is also evident by analyzing the
this. These compounds were compared to the active compound generated map of hydrophobic and hydrophilic fields for inhi-
within our original database, compound 25 using fingerprints bitor 25 (Fig. 8), where phenyl ring attached to thiadiazole and
based on the Tanimoto similarity metric as described in cinnamoyl moiety is buried in the hydrophobic pocket (orange
Materials and Methods section. The 2542 compounds with a color) while the ethylenic part is located in the hydrophilic
5016 M.F. Khan et al.

pocket (cyan color). This is in correlation with the 3D-QSAR important factors to be studied in concern with the absorption
result where hydrophobic R11 pharmacophoric feature (cin- of the drug molecule, which was further confirmed by pre-
namoyl moiety) and thiadiazole ring of 25 are found to be dicted Caco-2 cell permeability (QPPCaco), used as model
preferable for the activity (Fig. 7c). Our Glide XP-docking for gut-blood barrier. Caco-2 cell permeability prediction of
result also revealed solvent exposure in the region of thiadia- the test compounds indicates excellent results, predicting good
zole and cinnamoyl moiety plays an important role in sta- intestinal absorption. Further, the test results for QPlogkhsa
bilization of inhibitor at the active site. The compound also descriptor of Qikprop indicating the predicted values of
showed polar interactions with amino acid residues such as human serum albumin binding indicated that test molecules
Asn 350, Thr 314, Asn 258, Asn 349, Thr 240 and Thr 239. were found to fall within the permissible range (1.5 to 1.5).
Binding mode and 2D ligand interaction diagram of least Also, the Qikprop descriptor for blood/brain partition coeffi-
active compound of the series (45) are shown in Fig. 2 of Sup- cient QPlogBB showed reliable prediction for all the test com-
plementary Material. This compound showed polar interac- pounds and reference drugs. The cell permeability of the blood
tions with Thr 314, Asn 350, Asn 349, Asn 258 and Thr 353. brain barrier that mimics MDCK cells (QPPMDCK) also dis-
aa residues Met 259, Ile 378, Val 315, Leu 248, Leu 255 and played reliable results as they were well within the prescribed
some others were responsible for formation of hydrophobic range. The aqueous solubility parameter (QPlog S) of the test
bonds with the compound. The only positively charged residue entities was assessed and the compounds were also found to be
responsible for interactions was Lys 352. On the whole, extent in the permissible range (<0.5).
of interactions for least active compound (45) was much less in
comparison with that exhibited by the most active compound 4. Conclusion
(25).
In addition, the accuracy of the docking procedure was Combined computational approach was applied to give insight into the
determined by examining how closely the lowest energy poses structural basis and inhibition mechanism for a series of acyl 1,3,4-
(binding conformation) predicted by the object scoring func- thiadizole amide and sulfonamides as antitubulin agents. 3D-QSAR
tion, Glide score (GScore), which resembles an experimental modeling was performed to provide a structural framework for under-
binding mode as determined by X-ray crystallography. The standing the structure activity relationship of these compounds. The
root mean square deviation (RMSD) between the predicted atom based 3D-QSAR generated model AAHR.11 exhibited good cor-
relation and predictive power and satisfactory agreement between
conformation and the observed X-ray crystallographic confor-
experiment and theory. Molecular docking studies were performed
mation of colchicines (PDB Code: 1SA0) was found to be to produce possible binding poses for these compounds to tubulin.
0.853 Å (Fig. 7d), a value that suggests the reliability of Glide The low value of RMSD between the initial complex structure and
XP docking mode in reproducing the experimentally observed the energy minimized final average complex structure suggests that
binding mode of tubulin polymerization inhibitors and the the derived docked complex is close to the equilibrium. Further,
parameter set for Glide XP docking is reasonable to reproduce ADME predictions were performed for these compounds. Conclu-
the X-ray structure. Further, similar orientation was observed sively, the hits obtained on virtual screening of the database have pro-
between the superposition of conformation 25 best XP- vided new chemical starting points for design and development of
docking pose and 3D-QSAR pose (RMSD: 0.870 Å) (Fig. 9). novel tubulin targeting agents.

3.8. Lipinski’s rule for drug likeliness and in silico ADME Declaration of interest
prediction
This research did not receive any specific grant from funding
Different pharmacokinetic parameters of the compounds agencies in the public, commercial or not-for-profit sectors.
taken for the study were subjected to ADME predictions by
Qikprop v4.3. The compounds were assessed for their basic Acknowledgments
parameters of Lipinski’s rule of 5 and other pharmacokinetic
parameters. Table 8 (Supplementary Material) shows the The authors are thankful to Jamia Hamdard, New Delhi.
results obtained from Qikprop with their permissible range.
In general, an orally active compound should not have more Appendix A. Supplementary material
than 2 violations of the Lipinski rule. The active test com-
pounds in present study were not found violating the rule more
Supplementary data associated with this article can be found,
than the maximum permissible limits and thus proving their
in the online version, at http://dx.doi.org/10.1016/j.arabjc.
drug likeness properties.
2016.11.004.
The optimum values of the descriptors, polar surface area
and rotatable bonds also have great influence on oral bioavail-
ability of the drug molecules. The important parameters with References
their permissible ranges are delineated in Table 8 (Supplemen-
Abbas, S.H., Abuo-Rahma, A.A., Abdel-Aziz, M., Aly, O.M., Beshr,
tary Material). The optimum value of rotatable bonds (0–15)
E.A., Gamal-Eldeen, A.M., 2016. Synthesis, cytotoxic activity, and
and polar surface area (7–200 Å) holds a great importance tubulin polymerization inhibitory activity of new pyrrol-2(3H)-ones
on the oral bioavailability of the drug molecules. The active and pyridazin-3(2H)-ones. Bioorg. Chem. 66, 46–62.
test acyl 1,3,4-thiadizole amide and sulfonamides derivatives An, W., Wang, W., Yu, T., Zhang, Y., Miao, Z., Meng, T., Shen, J.,
demonstrated results of the descriptors, which were found to 2016. Discovery of novel 2-phenyl-imidazo[1,2-a]pyridine ana-
be within the prescribed range, thus owing to good bioavail- logues targeting tubulin polymerization as antiproliferative agents.
ability. Intestinal absorption or permeation is also one of the Eur. J. Med. Chem. 112, 367–372.
Acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents 5017

Cichero, E., Buffa, L., Fossa, P., 2011a. 3,4,5-Trisubstituted-1,2,4–4H- Kirchmair, J., Markt, P., Distinto, S., Wolber, G., Langer, T., 2008.
triazoles as WT and Y188L mutant HIV-1 non-nucleoside reverse Evaluation of the performance of 3D virtual screening protocols:
transcriptase inhibitors: docking-based CoMFA and CoMSIA RMSD comparisons, enrichment assessments, and decoy selec-
analyses. J. Mol. Model. 17, 1537–1550. tion—what can we learn from earlier mistakes? J. Comput. Aided
Cichero, E., Espinoza, S., Franchini, S., Guarenito, S., Brasili, L., Mol. Des. 22, 213–228.
Gainetdinov, R.R., Fossa, P., 2014. Further insights into the Li, D.D., Qin, Y.J., Zhang, X., Yin, Y., Zhu, H.L., Zhao, L.G., 2015.
pharmacology of the human trace amine-associated receptors: Combined molecular docking, 3D-QSAR, and pharmacophore
discovery of novel ligands for TAAR1 by a virtual screening model: design of novel tubulin polymerization Inhibitors by
approach. Chem. Biol. Drug Des. 84, 712–720. binding to colchicine-binding site. Chem. Biol. Drug. Des. 86,
Cichero, E., Espinoza, S., Tonelli, M., Franchini, S., Gerasimov, A.S., 731–745.
Sorbi, S., Gainetdinov, R.R., Brasili, L., Fossa, P., 2016. A Li, Y.J., Qin, Y.J., Makawana, J.A., Wang, Y.T., Zhang, Y.Q., Zhang,
homology modelling-driven study leading to the discovery of the Y.L., Yang, M.R., Jiang, A.Q., Zhu, H.L., 2014. Synthesis,
first mouse trace amine-associated receptor 5 (TAAR5) antago- biological evaluation and molecular modeling of 1,3,4-thiadiazol-
nists. Med. Chem. Comm. 7, 353–364. 2-amide derivatives as novel antitubulin agents. Bioorg. Med.
Cichero, E., Fossa, P., 2012. Docking-based 3D-QSAR analyses of Chem. 22, 4312–4322.
pyrazole derivatives as HIV-1 non-nucleoside reverse transcriptase Liao, S.Y., Chen, J.C., Miao, T.F., Shen, Y., Zheng, K.C., 2010.
inhibitors. J. Mol. Model. 18, 1573–1582. Binding conformations and QSAR of CA-4 analogs as tubulin
Cichero, E., Liqresti, A., Allara, M., Lazzati, Z., D’Ursi, P., inhibitors. J. Enzyme Inhib. Med. Chem. 25, 421–429.
Marabotti, A., Milanesi, L., Spallarossa, A., Ranise, A., Fossa, Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 2001.
P., 2011b. Homology modeling in tandem with 3D-QSAR analyses: Experimental and computational approaches to estimate solubility
a computational approach to depict the agonist binding site of the and permeability in drug discovery and development settings. Adv.
human CB2 receptor. Eur. J. Med. Chem. 46, 4489–4505. Drug Deliv. Rev. 46, 3–26.
Dixon, S.L., Smondyrev, A.M., Knoll, E.H., Rao, S.N., Shaw, D.E., Lokwani, D.K., Sarkate, A.P., Shinde, D.B., 2013. 3D-QSAR and
Friesner, R.A., 2006. PHASE: a new engine for pharmacophore docking studies of benzoyl urea derivatives as tubulin-binding
perception, 3D QSAR model development, and 3D database agents for antiproliferative activity. Med. Chem. Res. 22, 1415–
screening: 1. Methodology and preliminary results. J. Comput. 1425.
Aided Mol. Des. 20, 647–671. Luo, Y., Qiu, K.M., Lu, X., Liu, K., Fu, J., Zhu, H.L., 2011.
Dumontet, C., 2011. Antitubulin agents. B. Cancer 98, 1275–1285. Synthesis, biological evaluation, and molecular modeling of
Dumontet, C., Jordan, M.A., 2010. Microtubule binding agents: a cinnamic acyl sulfonamide derivatives as novel antitubulin agents.
dynamic field of cancer therapeutics. Nat. Rev. Drug Discov. 9, Bioorg. Med. Chem. 19, 4730–4738.
790–803. Mahindroo, N., Liou, J.P., Chang, J.Y., Hseih, H.P., 2006. Antitubu-
Dumontet, C., Sikic, B.I., 1999. Mechanisms of action of and lin agents for the treatment of cancer – a medicinal chemistry
resistance to antitubulin agents: microtubule dynamics, drug update. Expert Opin. Ther. Pat. 16, 647–691.
transport and cell death. J. Clin. Oncol. 17, 1061. Marzago, G., Chilin, A., 2014. QSAR and 3D-QSAR models in the
Friesner, R.A., Murphy, R.B., Repasky, M.P., Frye, L.L., Green- field of tubulin inhibitors as anticancer agents. Curr. Top. Med.
wood, J.R., Halgren, T.A., Sanschagrin, P.C., Mainz, D.T., 2006. Chem. 14, 2253–2262.
Extra precision glide: docking and scoring incorporating a model of Melagraki, G., Afantitis, A., 2013. Enalos KNIME nodes: exploring
hydrophobic enclosure for protein-ligand complexes. J. Comput. corrosion inhibition of steel in acidic medium. Chem. Intel. Lab.
Aided Mol. Des. 49, 6177–6196. Syst. 123, 9–14.
Golbraikh, A., Tropsha, A., 2002a. Beware of Q2! J. Mol. Gr. Model. Miller, M.D., Sheridan, R.P., Kearsley, S.K., 1999. SQ: a program for
20, 269–276. rapidly producing pharmacophorically relevant molecular Super-
Golbraikh, A., Tropsha, A., 2002b. Predictive QSAR modeling based positions. J. Med. Chem. 42, 1505–1514.
on diversity sampling of experimental datasets for the training and Mollinedo, F., Gajate, C., 2003. Microtubules, microtubule-interfering
test set selection. J. Comput. Aided Mol. Des. 5, 231–243. agents and apoptosis. Apoptosis 8, 413–450.
Hamel, E., 2003. Evaluation of antimitotic agents by quantitative Negi, A.S., Gautam, Y., Alam, S., Chanda, D., Luqman, S., Sarkar, J.,
comparisons of their effects on the polymerization of purified Khan, F., Konwar, R., 2015. Natural antitubulin agents: impor-
tubulin. Cell Biochem. Biophys. 38, 1–21. tance of 3,4,5-trimethoxyphenylfragment. Bioorg. Med. Chem. 23,
Hamel, E., Lin, C.M., 1984. Separation of active tubulin and 373–389.
microtubule-associated proteins by ultracentrifugation and isola- Nogales, E., Wolf, S.G., Downing, K.H., 1998. Structure of the ab
tion of a component causing the formation of microtubule bundles. tubulin dimer by electron crystallography. Nature 391, 199.
Biochemistry 23, 4173–4184. Sastry, G.M., Adzhigirey, M., Day, T., Annabhimoju, R., Sherman,
Jordan, A., Hadfield, J.A., Lawrence, N.J., McGown, A.T., 1998. W., 2013. Protein and ligand preparation: parameters, protocols
Tubulin as a target for anticancer drugs: agents which interact with and influence on virtual screening enrichments. J. Comput. Aided
the mitotic spindle. Med. Res. Rev. 18, 259–296. Mol. Des. 27, 221–234.
Kamal, A., Reddy, C.R., Vishnuvardhan, M.V.P.S., Mahesh, R., Sastry, G.M., Dixon, S.L., Sherman, W., 2011. Rapid shape-based
Nayak, V. Lakshma, Prabhakar, S., Reddy, C. Suresh, 2014. ligand alignment and virtual screening method based on atom/
Synthesis and biological evaluation of cinnamido linked ben- feature-pair similarities and volume overlap scoring. J. Chem.
zophenone hybrids as tubulin polymerization inhibitors and Inform. Model. 51, 2455–2466.
apoptosis inducing agents. Bioorg. Med. Chem. Lett. 24, 2309– Shearwin, K.E., Timasheff, S.N., 1994. Effect of colchicines analogs on
2314. the dissociation of ab tubulin into the subunits: the locus of
Kamal, A., Srikanth, P.S., Visdnuvardhan, M.V.P.S., Kumar, G.B., clchicine binding. Biochemistry 33, 894–901.
Babu, K.S., Hussaini, S.M.A., Kapure, J.S., Alarifi, A., 2016. Shen, M., Beguin, C., Golbraikh, A., Stables, J., Kohn, H., Tropsha,
Combretastatin linked 1,3,4-oxadiazole conjugates as potent tubu- A., 2004. Application of predictive QSAR models to database
lin polymerization inhibitors. Bioorg. Chem. 65, 126–136. mining: identification and experimental validation of novel anti-
Kandakatla, N., Ramakrishnan, G., Karthikeyan, J., Chekkara, R., convulsant compounds. J. Med. Chem. 47, 2356–2364.
2014. Pharmacophore modeling, atom based 3D-QSAR and Sheridan, R.P., Singh, S.B., Fluder, E.M., Kearsley, S.K., 2001.
docking studies of chalcone derivatives as tubulin inhibitors. Protocols for bridging the peptide to nonpeptide gap in topological
Orient. J. Chem. 30, 1083–1098. similarity searches. J. Chem. Inform. Comput. Sci. 41, 1395–1406.
5018 M.F. Khan et al.

Shivakumar, D., Williams, J., Wu, Y., Damm, W., Shelley, J., docking studies of cinnamic acyl 1,3,4-thiadiazole amide derivatives
Sherman, W., 2010. Prediction of absolute solvation free energies as novel antitubulin agents. Bioorg. Med. Chem. 20, 1181–1187.
using molecular dynamics free energy perturbation and the OPLS Zghaib, Z., Guichou, J.F., Vappiani, J., Bec, N., Hadj-Kaddour, K.,
force field. J. Chem. Theory Comput. 6, 1509–1519. Vincent, L.A., Paniagua-Gayraud, S., Larroque, C., Moarbess, G.,
Shobeiri, N., Rashedi, M., Mosaffa, F., Zarghi, A., Gahndadi, M., Cuq, P., Kassab, I., Deleuze-Masquefa, C., Diab-Assaf, M.,
Ghasemi, A., Ghodsi, R., 2016. Synthesis and biological evaluation Bonnet, P.A., 2016. New imidazoquinoxaline derivatives: synthesis,
of quinoline analogues of flavones as potential anticancer agents and biological evaluation on melanoma, effect on tubulin polymeriza-
tubulin polymerization inhibitors. Eur. J. Med. Chem. 114, 14–23. tion and structure-activity relationships. Bioorg. Med. Chem. 24,
Tropsha, A., Gramatica, P., Gombar, V.K., 2003. The importance of 2433–2440.
being earnest: validation is the absolute essential for successful Zhang, S., Golbraikh, A., Oloff, S., Kohn, H., Tropsha, A., 2006. A
application and interpretation of QSPR models. QSAR Comb. Sci. novel automated lazy learning QSAR (ALL-QSAR) approach:
22, 69–77. method development, applications, and virtual screening of chem-
Uppuluri, S., Knipling, L., Sackett, D.L., Wolff, J., 1993. Localization ical databases using validated ALL-QSAR models. J. Chem.
of the colchicines-binding site of tubulin. Proc. Natl. Acad. Sci. Inform. Model. 46, 1984–1995.
USA 90, 11598–11602. Zhou, J., Giannakakou, P., 2005. Targeting microtubules for cancer
Yang, X.H., Wen, Q., Zhao, T.T., Sun, J., Li, X., Xing, M., Lu, X., chemotherapy. Curr. Med. Chem. 5, 65–71.
Zhu, H.L., 2012. Synthesis, biological evaluation, and molecular

You might also like