Download as pdf or txt
Download as pdf or txt
You are on page 1of 28

Accepted Manuscript

Title: Clustering based drug-drug interaction networks for


possible repositioning of drugs against EGFR mutations:
Clustering based DDI networks for EGFR mutations

Authors: Anum Munir, Sana Elahi, Nayyer Masood

PII: S1476-9271(17)30871-X
DOI: https://doi.org/10.1016/j.compbiolchem.2018.04.011
Reference: CBAC 6843

To appear in: Computational Biology and Chemistry

Received date: 17-12-2017


Revised date: 30-3-2018
Accepted date: 15-4-2018

Please cite this article as: Munir, Anum, Elahi, Sana, Masood, Nayyer, Clustering
based drug-drug interaction networks for possible repositioning of drugs against EGFR
mutations: Clustering based DDI networks for EGFR mutations.Computational Biology
and Chemistry https://doi.org/10.1016/j.compbiolchem.2018.04.011

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
Clustering Based Drug-Drug Interaction Networks for Possible

Repositioning of Drugs against EGFR Mutations: Clustering

Based DDI Networks for EGFR Mutations

PT
Anum Munir1, 3*, Sana Elahi1, Nayyer Masood1,2

RI
1
Department of Bioinformatics and Biosciences, Capital University of Science and

SC
Technology, Islamabad Pakistan,

U
Department of Computer Science, Capital University of Science and Technology,

N
Islamabad Pakistan,
A
3
Bioinformatics International Research Club, Abbottabad, Pakistan
M

Corresponding Author: Anum Munir, Department of Bioinformatics and Biosciences,


D

Capital University of Science and Technology, Islamabad, 44000, Pakistan,


TE

Email: anummunir786@yahoo.com, Contact Number: +923348958178


EP
CC
A
Graphical abstract

PT
RI
SC
U
N
A
M
D
TE

Highlights:

 The strong interactions among the drugs are deemed as synergistic


EP

 The synergistic effect arises when the interactions cause an expansion in the effect
of either of the drug molecule.
CC

 The network structure is usually portrayed by a high modularity


 Modularity is directly connected to the links, which demonstrate the drug
A

interactions
 The centrality analysis is conducted to determine the strong interactions among the
drug compounds
 Toxic doses are frequently administered as LD50 values in mg/kg of the body
weight.
 The LD50 is the average lethal dose
 Different types of interactions are observed in each docked complex
 Drug-like properties based on Lipinski rule can be utilized as a part of the drug

PT
repurposing
 drug-like properties based on Lipinski rule can be utilized as a part of the new drug-

RI
drug interaction discoveries.

SC
U
Abstract
N
A
EGFRs are a vast group of receptor tyrosine kinases playing an important role in a number
M

of tumors, including lungs, head and neck, breast, and esophageal cancers. A couple of

techniques are being used in the process of drug design. Drug repositioning or repurposing
D

is a rising idea that consists of distinguishing modern remedial indications for officially
TE

existing dynamic pharmaceutical compounds. Here, a novel approach of analyzing drug-


EP

drug interaction networks, based on clustering methodology is used to reposition effective

compounds against EGFR mutations. Data about 2062 drugs are obtained, and mining is
CC

performed to filter only those drugs which fulfill Lipinski rule of five. Clustering is

performed, and DDIs are built on the clusters to identify effective drug compounds. Only
A

1052 compounds fulfill Lipinski rule. 12 clusters are formed for 1052 drugs compounds.

DDIs are developed for each cluster. Only 15 drugs are suggested to be more effective

assuming strong interactions in a DDI.


Keywords: Clustering, Drug-drug interaction, EGFR, Lipinski rule, Repositioning

Introduction

The epidermal Growth Factor receptor (EGFR) group of receptor tyrosine kinases (TKs),

PT
also known as the HER or ErbB family comprises four members HER1 (ErbB1), HER2

(ErbB2), HER3 (ErbB3) and HER4 (ErbB4) that direct numerous metabolic, physiological

RI
and developmental pathways (Gazdar, 2009) Stanley Cohen, a scientist, found EGFR 25

SC
years ago and explained its part in cell development (Seshacharyulu et al., 2012). The

EGFRs are a vast group of receptor tyrosine kinases playing an important role in a number

U
of tumors, including lungs, head and neck, breast, and esophageal cancers. EGFR and its
N
members are the significant contributors of signaling cascades, thus, regulate development,
A
signaling, migration, adhesion, development, and survival of tumor cells. Because of their
M

multidimensional role in a number of malignancies, EGFR family has risen as an important


D

candidate for the treatment of cancer (Grandis and Sok, 2004).


TE

Particularly the abnormal movement of EGFR has appeared to play a key role in the

development of tumor, where it is involved in the various cellular reactions, including


EP

apoptosis and cell proliferation (Wells, 1999). Nowadays, a lot of investigative studies are
CC

being done on EGFR ligand binding inhibition through the design of many antibodies and

therapies (Lin et al., 2003). A number of techniques are being used in the process of drug
A

design. Drug designing is a necessary piece of most medication disclosure programs

(Mountain, 2003) and is the real subject of research for some academic labs (Anderson,

2003). Cancer targets can be irritating because the targets are regularly physical cell
mutants of proteins that manage fundamental cell functions, bringing about the loss of a

general function. Obviously, it is irritating for a small inhibitor to potentiate the recovery of

a general function (Kaelin, 1999).

Drug repositioning or repurposing is a rising idea that comprises of distinguishing new

PT
remedial indications for officially existing dynamic pharmaceutical compounds (Udrescu et

RI
al., 2016; Sleigh et al., 2010; Munir et al., 2016). The rise of huge information and

examination has impelled the utilization of computational methodologies in numerous parts

SC
of pharmacology and medication configuration, including drug repurposing. In fact,

U
computational models are utilized to reveal drug interactions, which were not found during

N
clinical trials (Tatonetti et al., 2012), or to anticipate drug security (Egan et al., 2004).
A
Utilizing In-silico devices make a visual and natural framework for representing the
M

interactions (Lewis, 2010), in this manner helping medicinal and pharmaceutical practice.

A drug-drug interaction (DDI) is an unpredictable system in which the nodes represent


D

drugs, and the links between them correspond to the drug interaction, relationships (Polesk
TE

et al., 2011; Karlgren et al., 2011). Clustering methodologies are utilized to build DDI that
EP

proposes conceivable repositioning (Nugent et al., 2016). Here in this research work, the

same technique of clustering is applied to build the DDI to perform drug repositioning
CC

against the EGFR mutations. Though, current research proposes that interaction evidence

from DDI alone can be used to calculate physiological effects of the drug and therefore,
A

achieve drug repositioning

Material and Method


In this section, a novel procedure of drug repositioning against EGFR gene is proposed.

This procedure is based on the clustering of drug properties and development of DDIs.

First, the procedure of clustering is explained then the method of building DDIs and

repositioning of drugs based on DDIs is explained.

PT
Collection of Drugs Property Data

RI
The data of commercial compounds, which can be used in the treatment of EGFR

SC
mutations in various cancers, were obtained from Zinc database, through virtual screening.

The zinc is a database of commercial drug compounds; in addition to the chemical

U
structures of drug compounds, it also gives information about the drug-like properties of
N
the chemical compounds. Virtual screening is a computational method used in the parts of
A
drug innovation and improvement to discover collections of small ligands, which can
M

appropriately bind to their target proteins or enzymes (Raster, 2008; Rollinger et al., 2008).
D

The data is comprised of about 2052 records of the drug compounds, consisting of the
TE

chemical properties such as; Drug Zinc ID, Log P, Molecular weight (Mw), Hydrogen bond

donors(HBD), Hydrogen bond acceptors(HBA), Rotatable bonds (RB), A-polar


EP

dissociation and Polar dissociation.


CC

Mining of Data based on the Lipinski rule of five

Initially, the data consisted of all the drug's compounds, which can be used to treat EGFR
A

mutations. Most of the records lists contain obsolete drugs, which are toxic in nature and

have a lot of side effects. The data was sorted, and mining was performed to filter those

drug compounds that do not fulfill Lipinski rule of five and obtained only 1050 drugs
fulfilling the Lipinski rule of five. This rule demonstrates that drug-like compound must

have Mw no more than 500 Daltons, less than five HBD, less than ten HBA, and logP

ranges between 0 -5 (Gemma et al., 2006).

Clustering of Drug compounds

PT
Simple K means clustering was performed on the drug's information data in the Weka tool.

RI
WEKA is a prevalent machine learning work surface, contains applications of algorithms

SC
for clustering, classification, and association rule mining, along with the graphical user

interface and visualization services for data investigation and evaluation of an algorithm

U
(Bouckaert et al., 2010). K-means is one of the simplest unsupervised learning algorithms
N
used to solve the renowned clustering problem. This process follows a simple and easy way
A
to classify a given data set (x¬¬1, x2, x3…… xn) into a certain number of clusters, usually
M

K clusters, (K ≤ N) (Macqueen 1967). The optimal number of cluster K(n) was determined
D

using Elbow method given by Kodinariya and Makwana (2013) where K is the number of
TE

clusters and n is the data set.

Drug-Drug Interaction Network generation


EP

Drug-Drug interaction networks were built on the K clusters to determine the strong
CC

association between drug compounds within each cluster with the assistance of Gephi 0.9.1.

It is a principal tool for the visualization and analysis of large networks designed by
A

Andrew (2012). Each network consists of the set of vertices V and edge E, The average

path length L, The degree D of nodes, Network Density, and modularity classes. Modules
are particular parts of a system where the density of edges is fundamentally higher than

expected. For each network the L calculated by the formula

1
𝑙=
𝑛(𝑛 − 1)

PT
Where n is the number of vertices in the graph. The average clustering coefficient was

RI
calculated by the formula

SC
|𝐸𝑗, 𝑘 ∗ 𝑉𝑗, 𝑘𝑁|
𝐶=
𝐾𝑖(𝐾𝑖 − 1)

U
Where E is the edge, V is the vertex, N is the total number of nodes and Ki is the neighbor
N
vertices. The strong interactions among the drug compounds in each cluster were observed.
A
The modularity was calculated by the formula
M

𝑖 𝑑
𝑀 = Ʃ[ − ( ) ^2]
𝐸 2𝐸
D
TE

Where E is the number of edges in the network, i represent the number of strongly

connected edges, and d is the degree of a node. The drug compounds, which demonstrated
EP

stronger interactions in each cluster, were selected for further studies.


CC

Mining of strongly interacted drugs to select most suitable drugs

The strongly interacted drugs obtained through the DDI network were further analyzed for
A

their toxicity level, the drugs having lethal toxic classes were mined, and only those drugs

were selected, which demonstrate lower toxic classes.

Repositioning of strongly interacted drugs


The drug compounds, which demonstrated stronger interactions and low toxic classes, were

suggested to be used in combination with each other to achieve maximal results during the

treatment. DDIs reflect the intervention of several behaviors of the drug. In order to

understand potential repositioning, the strategy explained in this paper depends on the

PT
behavioral interactions between drugs rather than the structural similarities characterized by

RI
chemical-structures interactions or drug-target interactions. Therefore, to reduce the

complexity required by drug repurposing, it is suggested that this interactive perception can

SC
be integrated with the complementary structural perception (Bastian et al., 2009).

U
Results

Cluster Generation
N
A
M
The Drug property data obtained from the Zinc database were filtered based on a Lipinski

rule of five. Among the 2062 chemical compounds, only 1052 compounds fulfilled the
D

Lipinski rule of five and were selected for further analysis. By using the clustering
TE

methodology for grouping the similar chemical compounds, K-means algorithm was used.

12 K clusters were identified by Elbow method and built for 1052 drug compounds. The
EP

clusters were built for all the properties fulfilling Lipinski rule, shown in Fig 1. Each cluster
CC

contains only those drug compounds, which are similar to each other, based on their

properties, and are represented by different colors.


A

Development of Drug-drug Interaction networks

12 distinctive DDIs were built on the clustered data to find strong interacting drugs. In each

DDI every node represents a distinct drug. A connection between the two drugs relates to
the interaction between them according to the similarity of their properties. No data with

respect to the structural properties of these drug compounds is utilized throughout the

procedure. As each cluster contains similar drugs, therefore; the strong interactions among

the drugs demonstrate them as synergistic, because this type of interactions provides

PT
information about the functional profile of a drug cluster. At the point; when the

RI
interactions cause an expansion in the effect of either of the drug molecule the interaction is

known as a synergistic effect. An "additive synergy" happens when the last effect is

SC
equivalent to the sum of the effects of the two drugs. A point occurs when the final effect is

substantially more prominent than the sum of the two effects this is called enhanced

U
synergistic effect (Qato et al., 2016). The DDIs are shown in Fig 2.
N
A
The network 1 consists of 338 nodes and 562 edges; the network 2 is comprised of 205
M

nodes and 305 edges. Similarly; 373 nodes and 569 edges in network 3, 509nodes and 809

edges in network 4, 404 nodes and 592 edges in network 5, 238 nodes and 342 edges in
D

network 6, 424 nodes and 628 edges in network 7, 480 nodes and 799 edges in network 8,
TE

549 nodes and 914 edges in network 9, 328 nodes and 453 edges in network 10, 318 nodes
EP

and 428 edges in network 11, 470 nodes and 710 edges in network 12 were identified. It

was observed that network 9, 8, 4, and 12 contained maximum interacting drugs and edges
CC

as compared to other DDIs

Modularity and average clustering coefficient analysis


A

Modularity classes were calculated for each DDI. Distinct colors were assigned

automatically, to the nodes based on modularity. The network structure is usually portrayed
by a high modularity, both in the bipartite network and in their projections, showing that

topology is exceptionally distinct from an arbitrary network, and that it contains a rich and

heterogeneous modular structure (Zahoránszky-Köhalmi, 2016). The modularity based

coloring is steady. This outcome is a result of modularity being a decent indicator of

PT
properties and usefulness of chemical compounds (Qato et al., 2016). Additionally,

RI
modularity is directly connected to the links, which demonstrate drug interactions. The

average (Avg) degree, network density, modularity, Avg path length, and the Avg graph

SC
clustering coefficients are shown in Table 1.

U
Network Centrality Analysis

N
Centrality analysis was performed to determine the strong interactions among the drug
A
compounds, drugs that were most susceptible to DDIs link to the nodes with the largest
M

network centrality values were confirmed.


D

Repositioning of Strongly Interacted drugs


TE

From each DDI, drugs representing the strong interactions were selected, and a last drug-
EP

drug interaction network was built, based on the data consisting of strongly interacted drug

compounds. The last DDI is shown in Fig 3.


CC

The final DDI network contains 290 nodes and 416 edges. Again, the same procedure was
A

used to assign different colors to the nodes, average degree, network density, modularity,

average path length, and the average graph clustering coefficients were calculated and the

drug compounds that demonstrate strong interactions were selected. Finally, only 15 drugs
represent stronger interactions to each other. Values such as network density, clustering

coefficient, etc., of the final DDI network, are shown in Table 2.

The chemical properties of strongly interacted drugs found in the final DDI network,

fulfilling Lipinski rules of five are shown in table 3.

PT
The 15 strongly interacted results were further analyzed for the toxicity; Toxic doses are

RI
frequently administered as LD50 values in mg/kg of the body weight. The LD50 is the

SC
average lethal dose implying, the dose at which 50% of test subjects die upon introduction

to a compound. The drugs with fewer toxic values are shown in Table 4.

U
N
Only 11 drugs were lying in the toxicity class 4 and 5, both are non-toxic classes; Four
A
drugs were mined due to higher toxic values, based on results; these 11 strongly interacted
M
drugs are suggested to be used in the treatment of EGFR, and can be given to patients in

combination with each other to achieve maximal results of treatment.


D

This entire information presents the case for recouping various drug properties, for
TE

remaking some known drug repositioning, and the arrangements of conceivable drug-like
EP

properties based on Lipinski rule can be utilized as a part of the drug repurposing or in new

drug-drug interaction discoveries. This methodology can overcome numerous


CC

pharmacological issues as well as known repositioning, using only information about DDIs
A

Discussions

The clustering based, DDI network strategy introduced in this research work is portraying

drug interaction potential for the treatments of certain diseases. On the other hand, a few
parameters like modularity, path lengths, clustering coefficient, etc.., can distinguish those

drugs which have the most noteworthy potential for DDIs. By utilizing twofold clustering

techniques for DDI networks and drug repositioning, one can, undoubtedly, filter out useful

drug molecules from the jumble of drug compounds. There is a high likelihood that, the

PT
pharmacological properties by this twofold clustering strategy can be affirmed, these 15

RI
drug compounds can be additionally examined with in vivo and in vitro methods, to affirm

the conceivable repositioning or new interactions. The DDIs usually reflect the impedance

SC
of drug practices. The system clarified in this paper depends on the behavioral interactions

between drugs instead of the basic structural or drug target associations. It is proposed It is

U
proposed by Nacher and Schwartz (2012) that this behavioral point of view can be
N
coordinated with the structural perspective for better outcomes.
A
M

Nowadays, a lot of network-based methods are being developed; these methods have,

likewise, being utilized to distinguish drug candidates suitable for repositioning (Newman,
D

2003). For example, Chiang and Butte processed drug-drug similarity mechanism to
TE

recognize drug repositioning applicants (Zahoránszky et al., 2016), whereas some other
EP

used either drug-drug similarities (Wu et al., 2013; Chiang and Butte, 2011) or both drug-

drug and disease-disease similarity approaches (Yang and Agarwal, 2011; Cheng et al.,
CC

2012; Yutuka et al., 2013). Nonetheless, the vast majority of these methodologies is either

drug driven, disease driven or symptoms driven. Here in this research work, a DDI
A

approach is utilized based on the clustering strategy to recognize the best chemical

compounds, which can be utilized to treat EGFR mutations.


Several anticancer drugs have been created. Many patients with strong tumors still show the

poor prognosis. Hence it is critical to decide the proper utilization of such medicines

clinically. Several anti-cancer agents are being used. Various combinations of treatment

regimens have turned out to be effective (Assaf et al., 2011) and are broadly concerned

PT
with the introductory treatments for cancers (Keiser et al., 2009). However, at present, the

RI
effect of these treatments on enhancing persistent survival stays in a far away (Bonomi et

al., 2000; Schillar et al., 2002; Shepherd et al., 2013). Therefore, the method of identifying

SC
effective drugs by using DDIs based on clustering methods will be proved fruitful and will

show more satisfactory results.

U
Conclusion N
A
This approach to predict novel drugs by representing drug-drug interactions and using the
M

clustering methodology, not just prompted the proposal of drug repositioning, but
D

hopefully, permits additional knowledge into them. The predictions produced for a strongly
TE

interacted drug exhibit that this approach can viably recognize new indications and more

compelling drugs to treat EGFR mutations. 12 clusters were formed for 1052 drugs
EP

compounds. DDIs for developed for each cluster. Only 15 drugs are suggested to be more

effective based on strong interactions in a DDI and can be given in the combination to
CC

achieve maximal benefits. However, this is an In-silico study. Further examinations can be

performed in-vivo and in-vitro to confirm the efficacy of these suggested drug compounds.
A

Acknowledgments
The Authors are grateful to Bioinformatics International Research Club Abbottabad for

providing a platform to conduct research.

Conflict of Interests: None of the authors have any challenging conflict of interests

PT
References

RI
Anderson, A. C. (2003). The process of structure-based drug design. Chemistry & Biology,

Vol. 10 No. 9, pp. 787–797.

SC
Andrew Ng (2012). Clustering with the K-Means Algorithm, Machine Learning

U
Assaf G, Gideon YS, Eytan R, Roded S (2011). PREDICT: a method for inferring novel
N
drug indications with application to personalized medicine. Molecular Systems Biology,
A
Vol. 7, No. 1.
M

Bastian, M., Heymann, S., Jacomy, M. et al (2009). Gephi: an open source software for
D

exploring and manipulating networks. ICWSM, Vol. 8, pp. 361–362


TE

Bonomi P, Kim K, Fairclough D, Cella D, Kugler J, Rowinsky E, Jiroutek M, Johnson D


EP

(2000). Comparison of survival and quality of life in advanced non-small-cell lung cancer

patients treated with two dose levels of paclitaxel combined with cisplatin versus etoposide
CC

with cisplatin: Results of an Eastern Cooperative Oncology Group trial. J Clin Oncol, Vol.

18, pp. 623-631.


A

Bouckaert, R. R., Frank, E., Hall, M. A., Holmes, G., Pfahringer, B., Reutemann, P., &

Witten, I. H. (2010). WEKA−Experiences with a Java Open-Source Project. Journal of

Machine Learning Research, pp. 2533–2541.


Cheng F, Liu C, Jiang J, Lu W, Li W, Liu G, Zhou W, Huang J, Tang Y (2012). Prediction

of drug-target interactions and drug repositioning via network-based inference. PLoS

Comput Biol, Vol. 8, No. 5, e1002503.

Chiang AP, Butte AJ (2009). Systematic evaluation of drug-disease relationships to identify

PT
leads for novel drug uses. Clinical pharmacology and therapeutics, Vol. 86, No. 5, pp. 507-

RI
510.

SC
Egan, W. J., Zlokarnik, G. & Grootenhuis, P. D.(2004). In silico prediction of drug safety:

despite progress there is abundant room for improvement. Drug Discovery Today:

U
Technologies, Vol. 1, pp. 381–387
N
A
Gazdar, A. F. (2009). Activating and resistance mutations of EGFR in non-small-cell lung
M
cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene, Vol. 28, pp.

S24–S31. https://doi.org/10.1038/onc.2009.198
D

Gemma, A., Li, C., Sugiyama, Y., Matsuda, K., Seike, Y., Kosaihira, S., … Kudoh, S.
TE

(2006). Anticancer drug clustering in lung cancer based on gene expression profiles and
EP

sensitivity database. BMC Cancer, Vol. 6, No. 1. https://doi.org/10.1186/1471-2407-6-174

Grandis JR, Sok JC. (2004). Signaling through the epidermal growth factor receptor during
CC

the development of malignancy. Pharmacol Ther. Vol. 102, pp. 37–46. [PubMed:
A

15056497]
J. B. MacQueen (1967) "Some Methods for classification and Analysis of Multivariate

Observations, Proceedings of 5-th Berkeley Symposium on Mathematical Statistics and

Probability", Berkeley, University of California, Vol. 1, pp. 281-297

Karlgren, M. et al (2012). In vitro and in silico strategies to identify OATP1B1 inhibitors

PT
and predict clinical drug-drug interactions, Pharmaceutical research Vol. 29, pp. 411–426

RI
Kaelin, W. (1999). Choosing anticancer drug targets in the post- genomic era. J. Clin.

SC
Invest. Vol. 104, pp. 1503–1506.

U
Keiser MJ, Setola V, Irwin JJ, Laggner C, Abbas AI, Hufeisen SJ, Jensen NH, Kuijer MB,

N
Matos RC, Tran TB, Whaley R, Glennon RA, Hert J, Thomas KL, Edwards DD, Shoichet
A
BK, Roth BL(2009): Predicting new molecular targets for known drugs. Nature, Vol. 462,
M
No. 7270, pp. 175-181.

Kodinariya, T. M., & Makwana, P. R. (2013). Review on determining number of Cluster in


D

K-Means Clustering. International Journal, Vol. 1, No. 6, pp. 90–95.


TE

Lin, Q., Rana, P. ., Yingnian, L., Rajesh, A., Gail, H., Alex, F., & Michael, G. (2003).
EP

Epidermal Growth Factor Receptor mediates Silibinin - Induced Cytotoxicity in a Rat

Glioma Cell Line, Vol. 2, No. 5, pp. 526–531.


CC

Lewis, L. (2010). Drug-drug interactions: is there an optimal way to study them? British
A

journal of clinical pharmacology Vol. 70, pp. 781–783

Mountain, V. (2003). Astex, Structural Genomix, and Syrrx. Chem. Biol. Vol. 10, pp. 95–

98.
Munir, A., Azam, S., Fazal, S., Khan, Z., & Mehmood, A. (2016). In silico Repositioning

of Alendronate and Cytarabine Drugs to Cure Mutations of FPPS, HAP, PTPRS, PTPRE,

PTN4, GGPPS Gene and Mutant DNA, DPOLB, TOP2a, DPOLA, DNMT, RNA, TYSY,

RIR Genes. International Journal Bioautomation, Vol. 20, No. 3.

PT
Nacher, J. C., & Schwartz, J.-M. (2012). Modularity in Protein Complex and Drug

RI
Interactions Reveals New Polypharmacological Properties. PLoS ONE, Vol. 7, No. 1,

e30028. https://doi.org/10.1371/journal.pone.0030028

SC
Newman, M. E. (2003). The structure and function of complex networks. SIAM review

U
Vol. 45, pp. 167–256
N
A
Nugent, T., Plachouras, V. & Leidner, J. L. (2016). Computational drug repositioning based
M
on side-effects mined from social media. PeerJ Computer Science 2, e46

Polasek T, M., Lin F P, Y, Miners J, O., Doogue M, P. (2011). Perpetrators of


D

pharmacokinetic drug–drug interactions arising from altered cytochrome p450 activity: a


TE

criteria-based assessment. British journal of clinical pharmacology Vol. 71, pp. 727–736
EP

Qato DM, Wilder J, Schumm LP, Gillet V, Alexander GC (2016). "Changes in Prescription

and Over-the-Counter Medication and Dietary Supplement Use Among Older Adults in the
CC

United States, 2005 vs 2011". JAMA Internal Medicine. Vol. 176, No. 4, pp. 473–82.
A

doi:10.1001/jamainternmed.2015.8581
Rester U (2008). "From virtuality to reality - Virtual screening in lead discovery and lead

optimization: a medicinal chemistry perspective". Current Opinion in Drug Discovery &

Development. Vol. 11, No. 4, pp. 559–68. PMID 18600572.

Rollinger JM, Stuppner H, Langer T (2008). "Virtual screening for the discovery of

PT
bioactive natural products". Progress in Drug Research. Progress in Drug Research. Vol.

RI
65, No. 211, pp. 213–49.

SC
Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, Zhu J, Johnson DH

(2002). Comparison of four chemotherapy regimens for advanced non-small-cell lung

U
cancer. N Engl J Med, pp. 346:92-98.
N
A
Seshacharyulu, P., Ponnusamy, M. P., Haridas, D., Jain, M., Ganti, A. K., & Batra, S. K.
M
(2012). Targeting the EGFR signaling pathway in cancer therapy. Expert Opinion on

Therapeutic Targets, Vol. 16, No. 1, pp. 15–31.


D

https://doi.org/10.1517/14728222.2011.64861
TE

Shepherd FA, Dancey J, Ramlau R, Mattson K, Gralla R, O'Rourke M, Levitan N, Gressot


EP

L, Vincent M, Burkes R, Coughlin S, Kim Y, Berille J. (2010). Prospective randomized

trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer
CC

previously treated with platinum-based chemotherapy. J Clin Oncol, Vol. 2000, No. 18, pp.

2095-2103.
A

Sleigh, S. H. & Barton, C. L. (n.d.) Repurposing strategies for therapeutics. Pharmaceutical

Medicine Vol. 24, pp. 151–159


Tatonetti, N. P., Patrick, P. Y., Daneshjou, R. & Altman, R. B. (2012). Data-driven

prediction of drug effects and interactions. Science translational medicine Vol. 4, pp.

125ra31–125ra31

Udrescu, L., Sbârcea, L., Topîrceanu, A., Iovanovici, A., Kurunczi, L., Bogdan, P., &

PT
Udrescu, M. (2016). Clustering drug-drug interaction networks with energy model layouts:

RI
community analysis and drug repurposing. Scientific Reports, Vol. 6, No. 1.

https://doi.org/10.1038/srep32745

SC
Wells A.( (1999). EGF receptor. Int J Biochem Cell Biol. Vol. 31, pp. 637–43. [PubMed:

U
10404636]
N
A
Wu, C., Gudivada, R. C., Aronow, B. J., & Jegga, A. G. (2013). Computational drug
M
repositioning through heterogeneous network clustering. BMC Systems Biology, Vol. 7,

No. 5, S6.
D

Yang L, Agarwal P. (2011). Systematic Drug Repositioning Based on Clinical Side-


TE

Effects. PLoS ONE, Vol. 6, No. 12, :e28025.


EP

Yutaka Fukuoka DT, Hisamichi Ogawa. (2013). A two-step drug repositioning method

based on a protein-protein interaction network of genes shared by two diseases and the
CC

similarity of drugs. Bioinformation Vol. 9, No. 2, pp. 89-93.


A

Zahoránszky-Köhalmi, G., Bologa, C. G. & Oprea, T. I.(2016). Impact of similarity

threshold on the topology of molecular similarity networks and clustering outcomes.

Journal of cheminformatics, Vol. 8, No. 1.


PT
RI
SC
U
N
A
M
D
TE
EP
CC
A

Figure 1: The number of clusters formed by using every property of the 1052 drug compounds

Figure 2: Drug-drug interaction networks built for each cluster, small spheres represent drugs,

interactions between the drugs compounds are represented by dotted lines, whereas the strong
interactions are represented by solid lines. The strong interactions are represented by solid lines of

purple, green blue and orange color among the nodes.

Figure 3: The Drug-drug interaction network generated on strongly interacted drugs data. The

stronger interactions are shown by dark green color edges, which were selected for further analysis.

PT
RI
SC
U
N
A
M
D
TE
EP
CC
A
Table 1: Degrees, Densities, path lengths, and modularity values predicted for each Drug-drug

Interaction network

Average Network Graph Modularity Clustering Average

Degree diameter Density Coefficient Path

PT
length

RI
Network 3.213 5 0.005 0.587 0.021 0.9821

SC
Network 2.967 2 0.007 0.627 0.01 1.036

U
2

Network 3.051 2 0.004


N 0.59 0.001 1.039
A
3
M

Network 3.179 2 0.003 0.554 0.001 1.046

4
D

Network 2.931 2 0.004 0.598 0.002 1.036


TE

Network 2.874 2 0.006 0.617 0.001 1.063


EP

6
CC

Network 2.923 3 0.004 0.617 0.002 1.071

7
A

Network 3.329 2 0.003 0.595 0.002 1.047

Network 3.339 4 0.003 0.595 0.002 1.161


9

Network 2.762 1 0.004 0.61 0.021 1.0

10

Network 2.937 1 0.005 0.62 0.001 1.0

PT
11

RI
Network 3.021 2 0.003 0.595 0.002 1.095

12

SC
U
Table 2: Degrees, Densities, path lengths, and modularity values predicted for the final Drug-drug

N
Interaction network
A
Average Network Graph Modularity Clustering Average
M

Degree diameter Density Coefficient Path

length
D

Final 2.938 1 0.005 0.54 0.021 1


TE

Network
EP

Table 3: The Properties of the drug compounds, fulfilling Lipinski rule of five
CC

Selected Drugs XlogP Hydrogen Hydrogen Molecular Rotatable

bond donors bond weight bonds


A

acceptors

ZINC66252027 1.6 3 7 364 6

ZINC29128766 3.83 3 9 466 9


ZINC29043935 4.75 3 9 497 9

ZINC28903156 1.75 4 9 454 6

ZINC28903151 1.89 4 9 454 6

ZINC28903146 1.86 4 9 454 6

ZINC28817149 3.41 4 9 499 7

PT
ZINC29128814 3.19 4 9 426 9

RI
ZINC28901061 3.13 4 8 446 9

ZINC34642910 3.43 4 8 448 9

SC
ZINC34800033 3.47 4 8 461 9

ZINC64564748 4.41 4 7 472 9

U
ZINC29128958 3.54 4 9 493 9

ZINC26983604

ZINC29198958
1.56

1.53
5

3
N 9

7
397

365
9

7
A
M

Table 4: The drugs which lie in the lower toxic classes


D

S.no Structure Zinc ID Predicted Toxicity


TE

LD50 class

1
EP

ZINC66252027 1800 mg/kg Toxicity class

4
CC
A
2 ZINC29128766 1000 mg/kg Toxicity class

PT
3 ZINC29043935 1000 mg/kg Toxicity class

RI
SC
4 ZINC28903156 1000 mg/kg Toxicity class

U
4
N
A
M

5 ZINC28817149 2500 mg/kg Toxicity class

5
D
TE
EP

6 ZINC29128814 1000 mg/kg Toxicity class

4
CC
A
7 ZINC28901061 1600 mg/kg Toxicity class

PT
8 ZINC34642910 3160 mg/kg Toxicity class

RI
SC
9 ZINC64564748 1000 mg/kg Toxicity class

U
4
N
A
M

10 ZINC29128958 1000 mg/kg Toxicity class


D

4
TE
EP

11 ZINC29198958 5000 mg/kg Toxicity class


CC

5
A

You might also like