You are on page 1of 13

Journal of Molecular Structure 1224 (2021) 129006

Contents lists available at ScienceDirect

Journal of Molecular Structure


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

Computational identification of potential dipeptidyl peptidase (DPP)-IV


inhibitors: Structure based virtual screening, molecular dynamics
simulation and knowledge based SAR studies
Virendra Nath a, Manish Ramchandani a, Neeraj Kumar b, Rohini Agrawal c, Vipin Kumar a,∗
a
Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer-301817, India
b
Bhupal Nobles’ College of Pharmacy, Bhupal Nobles’University, Udaipur-313001, India
c
College of Pharmacy, JSS Academy of Technical Sciences, Noida, Uttar Pradesh

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

Article history: Type 2 Diabetes mellitus (T2DM) is a globally leading metabolic problem with increased morbidity and
Received 6 May 2020 mortality. Current medication therapies in the market to control diabetes are not sufficient and there-
Revised 13 July 2020
fore, there is further need to develop more selective and effective treatment approaches. Inhibition of
Accepted 31 July 2020
Dipeptidyl-peptidase-IV (DPP-IV) enzyme may serve as an interesting target for developing novel anti-
Available online 31 July 2020
diabetic drug candidate. In the present study, hierarchical virtual screening of drug like compounds was
Keywords: done followed by molecular dynamics simulation and knowledge-based structure-activity relation (SAR)
Binding energy study in order to retrieve hit compound as prospective inhibitors of DPP-IV enzyme. Important amino
Diabetes mellitus acid residues present in the active target site were acknowledged as vital and were also found to have
Docking similar interactions with the potential hits. Further, in silico technique was undertaken to identify ubiq-
DPP-IV uitous promising hits against DPP-IV enzyme and this was followed by calculation of binding energy
MD simulation
and absorption, distribution, metabolism, excretion (ADME) prediction that could possibly support their
SAR
pharmacokinetic prospective. Furthermore, stability study using molecular dynamics simulation of protein
complex was accomplished with the most capable targeted hit established in the present study. In the
end, comparative analysis of 3-dimensional binding pose, orientation and planar structure of the potential
retrieved hit was done with marketed drugs (alogliptin and sitagliptin) in order to develop knowledge-
based structure-activity relationship, which proved the successful designing of DPP-IV enzyme inhibitors.
© 2020 Published by Elsevier B.V.

1. Introduction zymes serve as their targets and the associated drug therapies
involved in Type2 DM involve Gulcagon like peptide (GLP-1), G-
Type2 diabetes mellitus is a worldwide rising public health con- Protein couple receptor40 (GPR40), G-Protein couple receptor120
cern. In U.S., more than 23 million people have a medically de- (GPR120), Sodium glucose transport-2 (SGLT-2), Peroxisome pro-
clared diabetes [1]. The World Health Organization (WHO) esti- liferator activated receptor-gamma (PPARγ ), etc. [3–7]. Among all
mates the number of people with diabetes to be approximately the targets associated in the mechanistic class for treatment of
180 million. This number is projected to double by 2030. Type 2 T2DM, Dipeptidyl-peptidase-4 (DPP-4) is a widely distributed ser-
diabetes mellitus (T2DM) is a progressive disease characterized by ine protease that is involved in glucose metabolism and is present
high levels of glucose resulting from insulin resistance and im- in solubilized form in blood or anchored into tissue membranes
pairment of insulin secretion [2]. Many of the proteins and en- and has also been proven as a validated target for antidiabetic
therapy recently. Further, inhibition of DPP-4 has been shown to
indirectly stimulate the secretion of insulin. The mechanism of
Abbreviations: ADME, Absorption Distribution Metabolism Excretion; HTVS, anti-diabetic therapy is probably through increased incretin (GLP-
High throughput virtual screening; MD, Molecular dynamics; MMGBSA, Molecu-
1 and GIP) release after the food intake, thus inhibiting release of
lar mechanics generalized born solvation accessible; PDB, Protein data bank; DPP,
Dipetidyl peptidase; RMSD, Root mean square deviation; RMSF, Root mean square
glucagon, which stimulates insulin secretion and thereby decreases
fluctuation; SAR, Structural activity relationship; SP, Simple Precision; T2DM, TypeII blood glucose levels [8,9]. Presently, some of the DPP-IV inhibitors
Diabetes mellitus; XP, Extra precision. are available worldwide such as; sitagliptin, vildagliptin, alogliptin,

Corresponding author at: Department of Pharmaceutical Sciences, Central Uni- linagliptin, teneligliptin, anagliptin, saxagliptin [10,11]. These sub-
versity of Haryana, Mahendergarh-123031, India.
strates are of different nature, in which alogliptin, tenegliptin,
E-mail address: vipbhardwaj@rediffmail.com (V. Kumar).

https://doi.org/10.1016/j.molstruc.2020.129006
0022-2860/© 2020 Published by Elsevier B.V.
2 V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006

Fig. 1. Overview of methodology in flow diagram.

sitagliptin, linagliptin are non-covalent, while saxagliptin and


vildagliptin are the covalent type of DPP-IV inhibitors [12]. Several
drugs having different chemical scaffolds were reported in litera-
ture against various diseases such as; quinoline are active as anti-
amoebic, antiviral, anti-parasitic, anti-microbial, anticancer, etc.
Oxadiazole acts as anti-mycobacterial and anti-tubercular agents
[13–15]. Similarly, In the mechanistic class of DPP-IV inhibitors;
quinazoline moiety in alogliptin, proline group in vildagliptin and
saxagliptin, adamantyl scaffold in vildagliptin have significant role
by forming hydrophobic contacts with amino acid residues in bind-
ing cavity of DPP-IV enzyme [16]. Crystal structure (PDB ID 2ONC)
data revealed good occupancy of the S1 pocket by these substrates
and it was also observed that they preferably reside inside the
hydrophobic cavity consisting of Arg125, Glu205, Glu206, Tyr547,
Tyr662, Tyr666, and Phe357 key residues [17,18].
The objective of the present study was to identify some novel
hits as DPP-IV inhibitors to develop future agents that could serve
as potential treatment option for type2 diabetes mellitus. Structure
based computational approach definitely reduces the expensive
experimental techniques and is also accepted widely in present
research scenario. Therefore, attempt has been made to identify
novel human DPP-IV inhibitors using molecular docking based vir-
tual screening, binding energy and ADME prediction on Chem- Fig. 2. Validation of Docking protocol.
bridge library of 15 lac compounds. Further, molecular dynamics
(MD) simulation studies of the DPP-IV (PDB ID 2ONC) with the
most promising hit molecule was undertaken to evaluate the sta- docking based virtual screening with good resolution of 2.55 Å. The
bility of these complexes and thus obtain the ligand-target selec- active site of 2ONC consists of an approved drug i.e. alogliptin [2].
tivity. To the best of our knowledge, this is the first example of The structure obtained from the PDB was prepared and mini-
successful application of both structure based virtual screening, mized using Protein Preparation Wizard of Schrodinger Drug Dis-
molecular dynamics simulation and knowledge based SAR studies covery suite 2017–2. This resulted in addition of hydrogens, cre-
for the identification of novel DPP-IV inhibitors. ation of disulfide bonds, assigning of proper bond orders, removal
of water molecules beyond 5Ǻ, optimization and minimization of
hydrogen bonds to remove any steric hindrances in the protein
2. Material and methodology structure. The filling of missing side chains and loops was done
by using Prime module [19].
2.1. Structure-based approach The prepared structures of human DPP-IV (2ONC) were used for
docking studies. 3D grid was created around the co-crystallized in-
The crystal structure of human dipeptidyl-peptidase-IV enzyme hibitor (alogliptin). Alogliptin binding site in receptor was found to
(hDPP-IV, PDB ID 2ONC) was obtained from protein databank for be located in central core of ligand binding domain which revealed
V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006 3

good occupancy of the S1 pocket by substrate and that, these drugs 2.2. Docking based virtual screening
reside inside with some key residues such as Arg125, Glu205,
Glu206, Tyr547, Tyr662, Tyr666, Ser630, and Phe357 [17,18]. The A library of 15 lac drug-like compounds was used to identify
size of grid was kept default at 20 Å. This site defines the binding the potential inhibitor of hDPP-IV. This chemical library was ac-
site of the hDPP-IV where molecular docking of the Chembridge quired from Chembridge database. This compound library was cho-
library of 15 lac ligand was performed. sen due to its diverse chemical moiety, thereby resulting in po-
Docking protocol was evaluated for robustness by extract- tential hits of diverse chemical scaffolds. Therefore, the screening
ing and preparing the co-crystalized inhibitor. The prepared ag- would be for the identification of novel scaffolds which could be
onist was then docked on the prepared grid of target using SP taken forward to hit optimization studies. All these chembridge lig-
docking technique implemented in GLIDE module version 6.9 of ands were prepared using LigPrep module version 3.6 for desalt-
Schrodinger Drug Discovery suite 2017–2. Root mean square de- ing, cleaning and addition of hydrogen atoms. Various stereoiso-
viation (RMSD) between the co-crystalized pose of the inhibitor mers, tautomers and ionization states at pH 7 ± 2 were also gen-
and the pose generated after SP docking was then calculated [20]. erated [21]. The 3D- grid was generated for docking based virtual
Smaller value of RMSD showed that the docking approach and screening at alogliptin binding site. The structure based drug de-
scoring function are able to mimic the crystal like pose which fur- sign, aimed to identify the most potent DPP-IV inhibitor and this
ther verified the correctness of the docking protocol. prepared library was subjected to molecular docking on the gener-

Fig. 3. Protein-ligand (PL) interactions; 3a. alogliptin; 3b. ZINC95497396; 3c. ZINC12432646; 3d ZINC72479286; 3e. ZINC12479967; 3f. ZINC95508153; 3 g. ZINC15078132.
4 V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006

Fig. 3. Continued

ated target grid using virtual screening workflow of GLIDE module 2.4. Binding energy calculations
version 6.9 (Hierarchal screening HTVS, SP, XP) in order to weed
out the molecules on the basis of shape complementarity of the The relative binding energy of selected agonists was calcu-
binding pocket [20]. lated from structural information in bimolecular complexes using
Prime module version 4.2 of Schrödinger software. Prime MM-
2.3. In silico ADME studies GBSA (Molecular Mechanics, the Generalized Born model and Sol-
vent Accessibility) is a physics-based method that computes the
The four major concerns in pharmacokinetics are absorption, force field energies in implicit solvent of the bound and unbound
distribution, metabolism and excretion (ADME). These four crite- molecules involved in the binding process. The MM-GBSA bind-
ria influence the drug levels and kinetics of drug exposure to the ing energy includes terms such as protein−ligand van der Waals
tissues and hence influence the performance and pharmacological contacts, electrostatic interactions, ligand desolvation, and internal
activity of the drug molecule. In silico pharmacokinetic properties strain (ligand and protein) energies. Prime calculates binding ener-
of the hits were predicted using QikProp module version 4.6 by gies using VSGB2.0 implicit solvent model with the OPLS2005 force
obeying rule of five and three in order to asses blood brain barrier field [24,25]. Retrieved hits obtained from docking on 2ONC were
permeability, polar surface area, human oral absorption percentage subjected to the MM-GBSA analysis individually. During these cal-
and molecular weight [22,23]. culations, the active site of protein was set to adjust itself up to
V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006 5

Fig. 3. Continued

a distance of 5 Å for ligand accordingly. This operation imports XP pared system of 2ONC-alogliptin and 2ONC-ZINC72479286 com-
docking “out.maegz” file of protein and ligand complex, which re- pleted for minimization step that contained 52,121 and 52,145
sults in ranking of the ligands based on the calculated binding en- atoms respectively. MD simulation of both complexes was per-
ergies [25,26]. formed for a period of 20 ns with a time-step of 2fs in NPT
(constant number of atoms N, pressure P, and temperature T)
2.5. Molecular dynamics simulation ensemble at 300 K temperature and 1 bar pressure. Tempera-
ture and pressure conditions were maintained by Nose−Hover
The structural stability of the protein-ligand complex was de- chain thermostat and Martyna−Tobias−Klein barostat, respec-
termined by molecular dynamics simulation of most promising tively. MD simulation was accomplished using multistep prac-
hit, i.e., ZINC72479286 and alogliptin with 2ONC. The 2ONC- tice present in Desmond software (version 3.8) [21,27,28]. Record-
alogliptin and 2ONC-ZINC72479286 complex were solvated using ing interval of 1.2 ps was used for energy and 4.8 ps were
explicit SPC (simple point charge) water molecules in orthorhom- used for trajectory for every complex. The structural stabil-
bic box. The system 2ONC-ZINC72479286 complex and 2ONC- ity and target-ligand interactions were analyzed over a pe-
alogliptin was neutralized by adding 67Na+ , 56Cl− and 68 Na+ , riod of 20 nanoseconds (ns) MD run. MD simulations in-
56 Cl− counterions respectively to stabilize the net charge of the cluded exhaustive screening workflow of methodology as shown
system. After addition of the solvent and counterions, the pre- in Fig. 1.
6 V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006

Fig. 3. Continued

Fig. 4. Root mean square deviation (RMSD) of protein ligand complex with both compounds during 20 ns of MD simulation; 4a. RMSD plot of 2ONC-Alogliptin; 4b. RMSD
plot of 2ONC- ZINC72479286.
V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006 7

Fig. 5. Root Mean Square Fluctuation (RMSF) of protein ligand complex with both compounds during 20 ns of MD simulation; 5a. RMSF plot of 2ONC-Alogliptin; 5b. RMSF
plot of 2ONC- ZINC72479286.

2.6. Knowledge based SAR studies ing (HTVS) followed by simple precision (SP) and the extra preci-
sion (XP). The crystal structure of human DPP-IV (PDB ID 2ONC)
Knowledge based SAR analysis was done with the help of FDA was chosen for target based virtual screening, and it clearly indi-
approved DPP-IV inhibitor alogliptin, sitagliptin and 3D structure of cated that the compounds having diversity in their structures were
hit. Docked orientation of hit compounds and approved drugs were able to form bonds with different amino acids in S1 pocket. The
overlapped in the binding pocket of human DPP-IV protein. Homo- active site of hDPP-IV was determined using co-crystallized ligand
geneous overlapping of compounds indicated the different features i.e. Alogliptin, which is the FDA approved drug (IC50 7 nM). Amino
that required for efficacy and affinity of hit compound [29,30]. acids which played key role for the inhibition of hDPP-IV enzyme
were mainly Glu 205, Glu 206, Arg 125, Tyr 547 and Try 631 of S1
2.7. Synthetic feasibility prediction pocket.
Using extra precision (XP) mode of docking, 132 compounds
The easiness of synthetic feasibility of selected hits was pre- were retrieved in hierarchal structure based virtual screening from
dicted by using Sylvia 1.4 server. Consideration of synthetic possi- the chosen Chembridge library of 15 lac diverse compounds. These
bilities was produced in a number between 1 and 10, lower num- compounds were obtained on the basis of affinity, pose, glide en-
ber indicate that simply synthesizable compounds, and higher for ergy, dock score and other calculated penalties between the target
compounds that are challenging to synthesize [31]. and ligand.

3. Results and discussion


3.3. Post processing hierarchical structure based screening
3.1. Validation of docking protocol
Weed-out processing included ADME prediction and binding
Self-docking of the bound ligand i.e. alogliptin in the 2ONC energy calculations. These screening processes of hits against
crystal structures of DPP-IV under optimized docking parameters hDPP-IV enzyme were done by using QikProp module version 4.6
gave RMSD of 2.011 Ǻ along with the dock score of −8.631. The of Schrodinger. Out of 15 lac compounds from chembridge library,
docked pose of co-crystal ligand is in orange colored ball stick only 06 potential hits were considered on the basis of different
shape and is overlapped with the co-crystal ligand which is in type of interactions obtained between enzyme and the ligand as
green colored ball stick model as shown in the Fig. 2. illustrated in Table 1. ADME predicted data and predictive bind-
ing energy are shown in Table 2. Based on similar interaction,
3.2. Structure based screening scores and admissible ADME, Compound 3 was selected for fur-
ther MD simulation studies as supported by the pose and orien-
The direct approach of screening was executed using hierarchal tation of compound 3 in binding cavity of 2ONC. Moreover, Com-
mode of elimination technique, i.e. high throughput virtual screen- pound 3 showed best results in 2D and 3D contacts with protein as
8 V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006

Fig. 6. Contacts of protein ligand complex with both compounds during 20 ns of MD simulation; 6a. 2D-contacts of 2ONC-Alogliptin; 6b. 2D-contacts 2ONC- ZINC72479286;
6c. Different interaction between 2ONC-Alogliptin; 6d Different interaction between 2ONC - ZINC72479286.

Table 1 Glu205, Glu206 showed hydrogen bonding in Alogliptin-DPP-IV


Protein-ligand (PL) interactions of drug and retrieved hits with their compound IDs.
contacts. These targeted compounds as depicted in Fig. 3 may
S. No. Compound ID P-L Interacting Residues serve as effective and potential hits for DPP-IV inhibition and this
Ref. Alogliptin Tyr666, Tyr547, Tyr631, Glu205, Glu206 process would also prove useful in lead finding process of drug de-
1 ZINC 95497396 Tyr666, Glu205, Glu206, Phe357 velopment procedure for diabetic population.
2 ZINC 12432646 Tyr666,Glu205, Glu206,Phe357, Ser209
3 ZINC 72479286 Tyr666, Glu205, Glu206, Phe357, Tyr547
4 ZINC 12479967 Tyr547, Tyr666, Glu206, Phe357
3.4. Molecular dynamics simulation
5 ZINC 95508153 Tyr666, Glu205, Glu206, Phe357, Arg125
6 ZINC 15078132 Arg125,Tyr662,Try666, Glu206
Diverse aspects were evaluated in MD simulation examination,
which mainly included the stability strength of protein-ligand by
analyzing root mean square deviation (RMSD) of backbone and
root mean square fluctuation of side chain residues. Molecular
compared to compound 1 and compound 2. Tyr547, Glu205, dynamics simulation studies of 2ONC with drug candidate, i.e.
Glu206 showed hydrogen bonding interactions and Tyr666 showed Alogliptin along with MD simulation of obtained best hit candidate
pi-pi bonding with compound 3 i.e. ZINC 72479286 whereas, (ZINC72479286) from exhaustive in silico screening against the tar-
Tyr547, Tyr666, Tyr662, Tyr631 showed pi-pi interaction while get were performed.
V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006 9

Fig. 6. Continued

3.4.1. MD simulation of complex hDPP-IV (2ONC) with Alogliptin and run. RMSF plot of side chains residues of protein showedleast
ZINC72479286 amount of fluctuations i.e. below 2 Ǻ as shown in Fig. 5(a)
Evaluation of the RMSD of the protein backbone with respect and (b). Protein- Ligand interactions that were maintained during
to the initial frame of protein structure was carried out so as to 20 ns MD simulation were examined. Residues ARG125, GLU205,
establish the stability of the protein complexes with bound in- GLU206, T YR547, T YR631, SER209, GLY632 and GLU204were in-
hibitor at the alogliptin binding site. Further, the structural sta- volved in interaction with alogliptin as shown in Fig. 6(a) and (c).
bility of the enzyme complexes was analyzed based on the RMSD Residues GLU205, GLU206, T YR547, T YR666,and PHE357 formed
and RMSF values of enzyme and ligands during the simulations. various interactions with ZINC72479286. TYR631, GLU206 and
The RMSD value of protein backbone was found to be stabilized GLU205 formed hydrogen bonded contacts while SER209, TYR547,
below 2 Ǻ. For the stable trajectories of both complexes i.e. in- GLY632 and GLU204 formed water bridges whereas TYR547 and
dividual alogliptin and ZINC72479286, RMSD were obtained be- ARG125showed pi-pi interactions. Residues GLU205 and GLU206
low 2.0 Ǻ as shown in Fig. 4(a) and (b). Binding energy of these were involved in hydrogen bonding interaction with ZINC72479286
ligands with protein was calculated using Prime MM-GBSA and as shown in Fig. 6(b) and (d). Residues T YR662, T YR666 and
was found to be −81.986 and −74.057 kcal/mol for alogliptin and HIS126 formed pi-pi stacking with ZINC72479286while residues
ZINC72479286 respectively. RMSF of both ligands with 2ONC com- PRO550 and TYR662 form contact through water bridge with
plexes were found to have stable trajectories during the 20 ns MD ZINC72479286.

Table 2
Dock score, Binding energy and ADME calculation.

S.No. ID Dock score Binding energy QPlogPo/w QPlogS QPlogHERG QPPCaco QPlogBB QPPMDCK PercentHumanOralAbsorption QPlogKp

1 95,497,396 −11.007 −83.978 2.582 −3.056 −5.898 320.861 −0.528 160.183 86.923 −4.012
2 12,432,646 −10.474 −84.966 2.955 −1.212 −6.331 135.692 −0.048 69.904 82.419 −4.882
3 72,479,286 −10.463 −74.057 3.484 −3.071 −7.136 172.125 0.494 90.395 87.36 −5.205
4 12,479,967 −10.378 −97.465 3.116 −2.73 −7.647 89.819 −0.056 69.06 80.151 −5.551
5 95,508,153 −10.417 −102.867 1.832 −1.051 −6.057 108.34 −0.133 54.806 74.089 −5.723
6 15,078,132 −9.183 −99.918 3.893 −2.527 −5.769 314.172 0.472 173.224 94.433 −5.009
10 V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006

Table 3
Depicting predicted synthetic accessibility score with their compound IDs and structures.

S.No. Compound ID Structures Synthetic accessibility score

Alogliptin 4.85

1 ZINC 95497396 4.01

2 ZINC 12432646 4.08

3 ZINC 72479286 3.69

4 ZINC 12479967 4.97

5 ZINC 95508153 4.29

6 ZINC 15078132 4.75


V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006 11

Fig. 7. Knowledge based SAR studies. a. Surface view of binding cavity with Alogliptin, Sitagliptin and ZINC72479286; b. 2D structures of drugs for assessment of important
pharmacophoric features in ZINC72479286.

3.4.2. Knowledge based SAR studies Modification with fused heterocyclic or bicyclic ring system
FDA approved drugs (alogliptin and sitagliptin) were used for with hetero atom in yellow region would be favored, electron
knowledge based SAR analysis and it was done by the evaluation withdrawing group(s) may be useful for substitution at green col-
of orientation and pose of these drugs in the binding pocket along ored region, the essentiality of orange colored site may be replace-
with overlapping of retrieved candidate compound. Coinciding of able from any cyclic ring or elongated carbon chain with or with-
these compounds (alogliptin is in red and sitagliptin is in yellow out heteroatom, purple region is essential for pi-pi interaction with
color) in the binding pocket revealed the fitness and shape com- key amino acids residues in the a binding pocket of DPP-IV en-
plementarity, which is considered essential for the affinity of hit zyme, therefore, insertion of aromatic system may produce a po-
candidate (purple color) and drugs. Further, 2D structural features tential inhibitor. The pink colored region is not essential for any
of alogliptin and sitagliptin was also matched with best obtained type of substitution as electron withdrawing group or donating
hit compound and it resulted in the similar chemical features of group were not involved in any kind of interactions as depicted
drugs and hit compound. in Fig. 7(a) and (b). Knowledge based SAR studies may be helpful
12 V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006

in designing and synthesis of novel DPP-IV inhibitors with more [2] J. Feng, Z. Zhang, M.B. Wallace, J.A. Stafford, S.W. Kaldor, D.B. Kassel, M. Navre,
selectivity and efficacy. L. Shi, R.J. Skene, T. Asakawa, K. Takeuchi, R. Xu, D.R. Webb, S.L. Gwaltney, Dis-
covery of alogliptin: a potent, selective, bioavailable, and efficacious inhibitor
of dipeptidyl peptidase IV, J. Med. Chem. 51 (2008) 4357. https://doi.org/10.
3.4.3. Sylvia—Synthetic accessibility estimation 1021/jm8006799.
[3] N. Tiwari, Therapeutic targets for diabetes mellitus: an update, Clin. Pharmacol.
The synthetic feasibility of retrieved hit compounds along with Biopharm. 3 (2014) 1–10. https://doi.org/10.4172/2167-065x.10 0 0117.
alogliptin had predicted scores as showed in Table 3. This finding [4] R.S. Bahare, G. Gupta, S. Malik, N. Sharma, New emerging targets for type-2
is similar to that of alogliptin that may be a valuable forward for diabetes, Int. J. Pharmtech. Res. 3 (2011) 809–818.
[5] N.D.T. Nguyen, L.T. Le, Targeted proteins for diabetes drug design, Adv. Nat. Sci.
the future synthesis with the feasible economic route. 3 (2012). https://doi.org/10.1088/2043-6262/3/1/013001.
[6] L.K.J. Stadler, I.S. Farooqi, A new drug target for type 2 diabetes, Cell 170 (2017)
12–14. https://doi.org/10.1016/j.cell.2017.06.024.
4. Conclusion [7] B.R. Miller, H. Nguyen, C.J. Hu, C. Lin, Q.T. Nguyen, New and emerging drugs
and targets for type 2 diabetes, Am. Health Drug Benefits 7 (2014). https://
The present study aimed to identify the best possible com- www.ncbi.nlm.nih.gov/pmc/articles/PMC4280522/pdf/ahdb- 07- 452.pdf.
[8] A. Barnett, DPP-4 inhibitors and their potential role in the management of
pounds as DPP-IV inhibitor from the chembridge library of type 2 diabetes, Int. J. Clin. Pract. 60 (2006) 1454–1470. https://doi.org/10.1111/
drug like compounds. Six thriving molecules were identified j.1742-1241.2006.01178.x.
using structure based drug design tools and it was observed [9] C. Schwehm, J. Li, H. Song, X. Hu, B. Kellam, M.J. Stocks, Synthesis of new DPP-
4 inhibitors based on a novel tricyclic scaffold, ACS Med. Chem. Lett. 6 (2015)
that among these six molecules, Compound 3 i.e. [(2-(3-((4-(1-
324–328. https://doi.org/10.1021/ml500503n.
methyl-1H-benzo[d]imidazol-2-yl) piperidin-1-yl)methyl)phenyl) [10] Y. Nakamura, Diabetes therapies in hemodialysis patients: dipeptidase-4 in-
ethanamine)] exhibited the best dock score, i.e. –10.463 K.cal/mol hibitors, World J. Diabetes 6 (2015) 840. https://doi.org/10.4239/wjd.v6.i6.840.
with DPP-IV (PDB ID-2ONC) and thus, was evaluated further for [11] B. Ahrén, DPP-4 inhibitors, best practice and research, Clin. Endocrinol. Metab.
21 (2007) 517–533. https://doi.org/10.1016/j.beem.2007.07.005.
protein-ligand interactions, ADME and predicted binding energy. [12] B. Ahrén, Emerging dipeptidyl peptidase-4 inhibitors for the treatment of di-
In addition, MD simulation was done to confirm the protein-ligand abetes, Expert Opin. Emerg. Drugs 13 (2008) 593–607. https://doi.org/10.1517/
complex stability. The RMSD of both, 2ONC with alogliptin and 14728210802584126.
[13] Y.S. Mary, P.B. Miniyar, Y.S. Mary, K.S. Resmi, C.Y. Panicker, S. Armaković, S.J. Ar-
compound 3 were found to be below 2.0 Å. Further, knowledge- maković, R. Thomas, B. Sureshkumar, Synthesis and spectroscopic study of
based SAR studies revealed the vital regions of binding pocket three new oxadiazole derivatives with detailed computational evaluation of
and thus, the diverse substitution sites in retrieved hits were their reactivity and pharmaceutical potential, J. Mol. Struct. 1173 (2018) 469–
480. https://doi.org/10.1016/j.molstruc.2018.07.026.
identified for the selection of more druggable compounds. These [14] M. Vraneš, A. Tot, S. Armaković, S. Armaković, S. Gadžurić, Structure making
retrieved compounds have cyclic or heteroaromatic ring which properties of 1-(2-hydroxylethyl)-3-methylimidazolium chloride ionic liquid, J.
formed hydrophobic contacts in the binding pocket of DPP-IV Chem. Thermodyn. 95 (2016) 174–179. https://doi.org/10.1016/j.jct.2015.12.012.
[15] V.V. Menon, E. Fazal, Y.S. Mary, C.Y. Panicker, S. Armaković, S.J. Armaković,
enzyme. Piperidine ring was present in sitagliptin and it was also S. Nagarajan, C. Van Alsenoy, FT-IR, FT-Raman and NMR characterization of 2-
present in the retrieved molecules 1, 3, 4, 5 and 6. Molecule 2 isopropyl-5-methylcyclohexyl quinoline-2-carboxylate and investigation of its
have proline moiety which was also present in vildagliptin and reactive and optoelectronic properties by molecular dynamics simulations and
DFT calculations, J. Mol. Struct. 1127 (2017) 124–137. https://doi.org/10.1016/j.
saxagliptin. Cyclized nitrile group was illustrated in molecule 3 as
molstruc.2016.07.096.
present in open form in alogliptin. Therefore, these compounds [16] V. Jha, K.S. Bhadoriya, Synthesis, pharmacological evaluation and molecular
may have inhibitory potential against the DPP-IV enzyme as docking studies of pyrimidinedione based DPP-4 inhibitors as antidiabetic
they have diversity in their structure and resemblance with the agents, J. Mol. Struct 1158 (2018) 96–105. https://doi.org/10.1016/j.molstruc.
2018.01.014.
previously approved drugs. The predicted synthetic accessibility [17] Z. Zhang, M.B. Wallace, J. Feng, J.A. Stafford, R.J. Skene, L. Shi, B. Lee, K. Aert-
score also proved the ease of synthetic feasibility of compound 3 geerts, A. Jennings, R. Xu, D.B. Kassel, S.W. Kaldor, M. Navre, D.R. Webb,
as compared to the other hit candidates. The results obtained in S.L. Gwaltney, Design and synthesis of pyrimidinone and pyrimidinedione in-
hibitors of dipeptidyl peptidase IV, J. Med. Chem. 54 (2011) 510–524. https:
the present study support the design of newer DPP-IV inhibitors //doi.org/10.1021/jm101016w.
as anti-diabetic lead compound. [18] S. Arulmozhiraja, N. Matsuo, E. Ishitsubo, S. Okazaki, H. Shimano, H. Tokiwa,
Comparative binding analysis of dipeptidyl peptidase IV (DPP-4) with antidia-
betic drugs -an ab initio fragment molecular orbital study, Plos One 11 (2016)
Declaration of Competing Interest 1–15. https://doi.org/10.1371/journal.pone.0166275.
[19] G. Madhavi Sastry, M. Adzhigirey, T. Day, R. Annabhimoju, W. Sherman, Pro-
tein and ligand preparation: parameters, protocols, and influence on virtual
The authors declare that they have no known competing finan- screening enrichments, J. Comput. Aided Mol. Des. 27 (2013) 221–234. https:
cial interests or personal relationships that could have appeared to //doi.org/10.1007/s10822- 013- 9644- 8.
influence the work reported in this paper. [20] R.C.Æ.D. Joseph-mccarthy, Virtual fragment screening : an exploration of vari-
ous docking and scoring protocols for fragments using Glide, (2009) 527–539.
https://doi.org/10.1007/s10822- 009- 9281- 4.
CRediT authorship contribution statement [21] S. Sharma, S. Srivastava, A. Shrivastava, R. Malik, F. Almalki, K. Saifullah,
M.M. Alam, M. Shaqiquzzaman, S. Ali, M. Akhter, Mining of potential dipeptidyl
peptidase-IV inhibitors as anti-diabetic agents using integrated in silico ap-
Virendra Nath: Conceptualization, Methodology, Validation, proaches, J. Biomol. Struct. Dyn. (2019). https://doi.org/10.1080/07391102.2019.
Data curation, Writing - original draft. Manish Ramchandani: Data 1701553.
[22] A. Chatterjee, S.J. Cutler, R.J. Doerksen, I.A. Khan, J.S. Williamson, Discovery of
curation, Visualization. Neeraj Kumar: Validation, Visualization.
thienoquinolone derivatives as selective and ATP non-competitive CDK5/p25
Rohini Agrawal: Writing - review & editing. Vipin Kumar: Project inhibitors by structure-based virtual screening, Bioorg. Med. Chem. 22 (2014)
administration, Resources. 6409–6421. https://doi.org/10.1016/j.bmc.2014.09.043.
[23] C.D. Dzierba, A.J. Tebben, R.G. Wilde, A.G. Takvorian, M. Rafalski, P. Kasireddy-
Polam, J.D. Klaczkiewicz, A.D. Pechulis, A.L. Davis, M.P. Sweet, A.M. Woo,
Acknowledgement Z. Yang, S.M. Ebeltoft, T.F. Molski, G. Zhang, R.C. Zaczek, G.L. Trainor,
A.P. Combs, P.J. Gilligan, Dihydropyridopyrazinones and dihydropteridinones
as corticotropin-releasing factor-1 receptor antagonists: structure-activity re-
I am thankful for the funding as Senior Research Fellow grant lationships and computational modeling, J. Med. Chem. 50 (2007) 2269–2272.
from Council of Scientific and Industrial Research, Government of https://doi.org/10.1021/jm0611410.
India (File no. 09/1131(0027)/19-EMR-I). [24] S. Sirin, R. Kumar, C. Martinez, M.J. Karmilowicz, P. Ghosh, Y.A. Abramov, V.
Martin, W. Sherman, A computational approach to enzyme design : predicting
ω - aminotransferase catalytic activity using docking and MM-GBSA scoring,
References (2014).
[25] S. Bhowmick, N.A. Alfaris, J.Z. Altamimi, Z.A. Alothman, T.S. Aldayel,
[1] R. Pathak, M.B. Bridgeman, Dipeptidyl peptidase-4 (DPP-4) inhibitors in the S.M. Wabaidur, A. Islam, Screening and analysis of bioactive food compounds
management of diabetes, P T 35 (2010) 509–513. for modulating the CDK2 protein for cell cycle arrest: multi-cheminformatics
V. Nath, M. Ramchandani and N. Kumar et al. / Journal of Molecular Structure 1224 (2021) 129006 13

approaches for anticancer therapeutics, J. Mol. Struct. (2020) 128316. https: [29] T.A. Halgren, Identifying and characterizing binding sites and assessing drug-
//doi.org/10.1016/j.molstruc.2020.128316. gability, J. Chem. Inf. Model. 49 (2009) 377–389. https://doi.org/10.1021/
[26] S. Gupta, D. Parihar, M. Shah, S. Yadav, H. Managori, S. Bhowmick, P.C. Patil, ci800324m.
S.A. Alissa, S.M. Wabaidur, M.A. Islam, Computational screening of promising [30] V. Nath, R. Agrawal, V. Kumar, Structure based docking and molecular dynam-
beta-secretase 1 inhibitors through multi-step molecular docking and molecu- ics studies: peroxisome proliferator-activated receptors –α /γ dual agonists for
lar dynamics simulations - pharmacoinformatics approach, J. Mol. Struct. 1205 treatment of metabolic disorders, J. Biomol. Struct. Dyn. 38 (2020) 511–523.
(2020) 127660. https://doi.org/10.1016/j.molstruc.2019.127660. https://doi.org/10.1080/07391102.2019.1581089.
[27] W. Humphrey, A. Dalke, K. Schulten, VMD: Visual Molecular Dynamics 7855 [31] K. Boda, T. Seidel, J. Gasteiger, Structure and reaction based evaluation of
(1996) 33–38. synthetic accessibility, J. Comput. Aided Mol. Des. 21 (2007) 311–325. https:
[28] D.E. Shaw, Atomic-level characterization, Science 341 (2012) 341–346. https: //doi.org/10.1007/s10822- 006- 9099- 2.
//doi.org/10.1126/science.1187409.

You might also like