143
Al-Azhar UniversityGaza Deanship of Postgraduate Studies Faculty of Pharmacy Master of Pharmaceutical Sciences Computational Approach Towards Exploring the Polypharmacology of Urtica dioica and Salvia officinalis Focusing on Antidiabetic, Antihyperlipidemic and Anticancer Activities By Asmaa Mahmoud Rabie Rabie Supervisor Prof. Dr. Ihab Almasri Prof. Dr. of Medicinal Chemistry & Drug Discovery A Thesis Submitted in Partial Fulfillment of the Requirements for the Degree of Master in Pharmaceutical Sciences January-2021

Computational Approach Towards Exploring the

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Computational Approach Towards Exploring the

Al-Azhar University–Gaza

Deanship of Postgraduate Studies

Faculty of Pharmacy

Master of Pharmaceutical Sciences

Computational Approach Towards Exploring the

Polypharmacology of Urtica dioica and Salvia officinalis Focusing on

Antidiabetic, Antihyperlipidemic and Anticancer Activities

By

Asmaa Mahmoud Rabie Rabie

Supervisor

Prof. Dr. Ihab Almasri

Prof. Dr. of Medicinal Chemistry & Drug Discovery

A Thesis Submitted in Partial Fulfillment of the Requirements for the Degree of

Master in Pharmaceutical Sciences

January-2021

Page 2: Computational Approach Towards Exploring the
Page 3: Computational Approach Towards Exploring the

i

Declaration

I declare that the thesis titled ―Computational Approach Towards Exploring the

Polypharmacology of Urtica dioica and Salvia officinalis Focusing on

Antidiabetic, Antihyperlipidemic and Anticancer Activities‖ submitted for the

degree of master of pharmaceutical sciences, is the result of my own research work

and the work provided in this thesis, unless otherwise referenced, is my own work,

and has never been submitted elsewhere for any other degree qualifications neither

for any academic titles, nor for any other academic or publishing institutions.

I declare that I will be responsible for academic and legal terms if this work

proves the opposite.

Signature:

Asmaa M R Rabie

Date: 20/1/2021

Page 4: Computational Approach Towards Exploring the

ii

Dedication

To my great parents, who never stop giving of themselves in countless ways,

To my dearest husband, who leads me through the valley of darkness with light

of hope and support,

To my beloved kids: Moath, Haya, Hala and Mohammed whom I can't force

myself to stop loving,

To my brothers and sisters, the symbol of love, generous and giving,

To my friends who encourage and support me,

To all the people in my life who touch my heart,

Asmaa M R Rabie

Page 5: Computational Approach Towards Exploring the

iii

Acknowledgment

In the Name of Allah, the Most Merciful, the Most Compassionate all praise be to Allah, the

Lord of the worlds; and prayers and peace be upon Mohamed His servant and messenger.

First and foremost, all my prayers and my limitless thanks to Allah, the Ever-Magnificent; the

Ever-Thankful, for His help and bless. I am totally sure that this work would have never become

truth, without His guidance.

I would like to express my profound gratitude and deep regard to my supervisor, Prof.Dr. Ihab

Almasri for his improving suggestions, advices, guidance, patience and constant encouragement

throughout this thesis, so I ask Allah to reward him on my behalf.

I owe a deep debt of gratitude to our university, Al-Azhar University, Gaza, for giving us an

opportunity to complete this work. I extend my appreciation to Faculty of Pharmacy and to all

the academic staff in it.

Thanks to the OpenEye Scientific company for Omega 2.5.1.4, & OEDOCKING 3.2.0.2

Softwares donation to Al-Azhar University, which were used in the study.

I owe profound gratitude to my husband, Raed, whose constant encouragement, limitless giving,

patience and great sacrifice, helped me accomplish my degree. Without his support, this study

would not have been possible.

My sincere thanks to my colleagues, pharmacy staff members and partner in working field in

Ministry of Health, especially in Shohadaa Jabalia Martyrs Health center as well as in Hala El-

Shawa Health Center,

Finally, my appreciation goes to all my beloved friends for their encouragement and support

along the way of doing my thesis.

Page 6: Computational Approach Towards Exploring the

iv

Abstract

Natural plants have long been considered as the cornerstones of drug discovery and

development. They have a wide range of diversity of multidimensional chemical structures; in

the meantime, the utility of natural products as broad biological target modifiers has also gained

considerable attention. Therefore, the identification of the molecular targets of natural

compounds is a critical step in rational design of more selective, potent and safer drugs. In this

work, we explored the polypharmacology of S. Officinalis and U. dioica and their

phytochemicals focusing on anticancer, antidiabetic and antihyperlipidemic activities using a

ligand-based target fishing approach. The fishing protocol was started with the generation of a

chemogenomic database that links individual targets with specific target ligands or group of

drugs. Targets profile was then generated using ROCS software. The applied method was able to

retrieve known on-targets as we had found that, many of our natural constituents could bind to

ER, MAPK14, PIK3CG and PPAR-γ such as apigenin, luteolin, oleanolic acid and quercetin.

The applied method was also able to identify potential off-targets. The validity of these off-

targets as potential targets were evaluated by docking simulation according to our adopted

experimental procedures, which include preparation of proteins, preparation of ligands and then

docking using FRED software version 3.3.1.2 (FRED, 2015) within the OEDockind suite in the

presence of the explicit water molecules. The obtained results clarified that the different

phytochemicals of S. Officinalis and U. dioica were successfully docked within the active sites of

the off-targets with relatively good scores such as DAPK1, Tank2, CDK2, CK2 and FGFR1.

These off-targets were consistent with recently identified bioactivities of S. Officinalis and U.

dioica and their phytochemicals.

In our results, the concept of polypharmacology is obviously clarified as multiple

phytochemicals could affect the same target on the disease pathway producing synergistic effect

or an individual compound could affect multiple targets that involved in the same disease.

Finally, further in in-vitro and in-vivo studies on the phytochemicals of S. officinalis and U.

dioica against the fished off-targets will provide more understanding of their pharmacological

properties as well as could provide good leads for designing new more potent and safer

anticancer, dantidiabetic and hypolipidemic drugs.

Keywords: Natural plants; Salvia officinalis; Urtica dioica; Polypharmacology; Similarity

search; Target fishing; Docking.

Page 7: Computational Approach Towards Exploring the

v

Abstract in Arabic

الملخص باللغة العربية

اليياشررا الميسياةيرر تشره مرر ياسردى سررفيررا تتسر دويةرر يتطهةرىرا فررا اشتذرال اأ ساسري الركيررزا اأ ت الطبيعير شباترراال تعتبرريعد تحديرد ك،ياس اىتسام كبير لذل حيهة على نطاقدتقبلات كسالشباتات الطبيعي اأ عادو ؛ فا نفس الهقت، حظيت متعددوا

خطررها حاسررس فررا الترررسيي العكلانررا لسركبررات أشوررر انتكاةيرر يفعاليرر يأمان ررا فررا ىررذا شباتررات الطبيعيرر للالجزةئيرر السدررتقبلاتيالسرهادو الميسياةير الشباتير الخابر يسرا مر التركيرز ي الكررة السرمير ات الدياةير الستعرددوا لشبتترا قسشرا اشتذرال الترر ير ،العسا

عسرا الجديردا للسركبرات ليرات الآ التعررل علرى نرةكر فرر حرحسيات الردم اسرتخدامي نذط السزرادوا للدررنان يالدر ر على اأ سدرتقبلاتات كيسياةي جيشهمير ترر ا القاعدا يان إنذاء بريتهكهل ىذا ال دأ تسادوا على مركبات ذات فعالي معريف الطبيعي اعكررد ل ((ROCS اسررتخدام رنررام السدررتقبلاتمحررددوا أي مجسهعرر مرر اأدويةرر رري ترري رنذرراء ملرر تعرةرر سدررتقبلاتالفردويرر حير يجردنا أن الس هنرات الطبيعير السختلفر السعريفر للسركبرات الطبيعير ى تحديرد السدرتقبلاتالطرةك السطبكر قرادو ا علرشانت ,ER مورررا اأ جيشررري ، اللهتيرررهلي ، حسررر اأيليانهلرررك يالمهةرسرررتي قرررادو ا علرررى ات تبرررا سدرررتقبلات معريفررر مترررا يللشبتتررر

MAPK14, PIK3CG وPPAR-γ ) أدوت أيزا الى التعررل علرى مدرتقبلات لري تمر معريفر مر قبرا الطرةك السطبك ،جرراءات تجر تشرا السعتسردا الترا نرةكر محاشراا اس سراء حدر ركييي فعالي ىذه السدتقبلات كسدتقبلات محتسلر اسرتخدام ي تي ت

أن ةي أفرادوت الشترا ) 3 3 1 1( نبعر FRED رنام ساء استخدام السركبات ي تطبيق اسشات ي تتزس تحزير البريتي ,DAPK1مورا مهاق ات تبا م السدتقبلات الجديدا ساء دواخااسقادو ا على كانت كرة ي ال السرمي العديد م مركبات

Tank2, CDK2, CK2وFGFR1 متهافكر مر اأنذرط الحيهةر الترا تري تحديردىا مر خر ا ( كسرا أن ىرذه السدرتقبلات الجديردا ي الكرة يالسهادو الميسياةي الشباتي الخاب يسا السرمي لشبتتا

جليا، حي تحظشا عدا مركبات م نفرس الشبتر تر ر علرى أحرد السدرتقبلات تعددو التر يرات الدياةي البح يظير مبدأ نتاة فا على السرض أي أن مرك ياحد ي ر على عدا مدتقبلات للسرض مسا يعطا تر يرا مزاعفا للسركبات الطبيعي التا ت ر

علررى السركبرات الطبيعيرر للسراميرا ي الكرررة ي السدرربقبلات يفرا اأجدررام الحيهةر أخيررا، السزةررد مر الد اسررات العسلير السخبرةرر كسرا سريهفر مركبرات جيردا لتررسيي أدويةر ليرذه السركبرات، الدياةير الجديدا التا تي اشتذافيا سهل ي دو للسزةد م فيي الخهاص

ي ضد الدرنان يالد ر ي فر ححهم الدممانا ي فعالأشور أ

لة:الكلمات الدا

ساء، اسسدتقبلاتدياةي ، ح التذا و، بيد ال، الكرة ، تعددو التر يرات الالسرمي الطبيعي ، شباتاتال

Page 8: Computational Approach Towards Exploring the

vi

List of contents

Declaration .................................................................................................................................. i

Dedication .................................................................................................................................. ii

Acknowledgment....................................................................................................................... iii

Abstract ..................................................................................................................................... iv

Abstract in Arabic ...................................................................................................................... v

List of contents .......................................................................................................................... vi

List of Tables ............................................................................................................................. x

List of Figures .......................................................................................................................... xii

List of Abbreviations ............................................................................................................... xiv

Chapter One Introduction ........................................................................................................... 1

1.1 Background ..................................................................................................................... 1

1.2 Problem statement ........................................................................................................... 2

1.3 Aim ................................................................................................................................. 2

4.1 Objectives ....................................................................................................................... 3

1.5 Justification of the Study ................................................................................................. 3

Chapter Two Literature Review .................................................................................................. 4

2.1 Drug discovery and importance of medicinal plants ........................................................ 4

2.2 Sage ................................................................................................................................ 5

2.2.1 Ethnomedicinal uses of S. officinalis ........................................................................ 6

2.2.2 Phytochemical constituents of S. officinalis .............................................................. 6

2.2.3 Pharmacological activities of S. officinalis ................................................................ 8

2.2.3.1 Anticancer effects .............................................................................................. 8

2.2.2.2 Antidiabetic effects ............................................................................................ 9

2.2.3.3 Antihyperlipidemic effects................................................................................. 9

2.2.3.4 Other effects ...................................................................................................... 9

2.2.4 Clinical studies ......................................................................................................... 9

2.3 Stinging Nettle .............................................................................................................. 10

2.3.1 Ethnomedicinal uses of U. dioica ........................................................................... 11

2.3.2 Phytochemical constituents of U. dioica ................................................................. 11

Page 9: Computational Approach Towards Exploring the

vii

2.3.3 Pharmacological activities of U. dioica ................................................................... 14

2.3.3.1 Antidiabetic effects .......................................................................................... 14

2.3.3.2 Antihyperlipidemic effects .............................................................................. 15

2.3.3.3 Anticancer effects ............................................................................................ 15

2.3.3.4 Other effects .................................................................................................... 15

2.4 Cancer .......................................................................................................................... 16

2.4.1 Causes & risk factors of cancer............................................................................... 16

2.4.2 Oxidative stress and cancer..................................................................................... 17

2.4.3 Signs and symptoms ............................................................................................... 18

2.4.4 Cancer treatment .................................................................................................... 18

2.4.4.1 Cancer treatment by drugs ............................................................................... 18

2.4.4.2 Cancer treatment by natural plants ................................................................... 20

2.5 Diabetes mellitus (D.M) ................................................................................................ 22

2.5.1 Classification of diabetes mellitus .......................................................................... 22

2.5.2 Signs and symptoms ............................................................................................... 23

2.5.3 Complications of diabetes mellitus ......................................................................... 23

2.5.4 T2DM pathogenesis and major risk factors ............................................................. 23

2.5.5 Management of T2DM ........................................................................................... 25

2.5.5.4 Non pharmacological treatment ....................................................................... 25

2.5.5.2 Pharmacological treatment............................................................................... 25

2.5.5.3 Diabetes management by natural plants ........................................................... 26

2.6 Hyperlipidemia ............................................................................................................. 27

2.6.1 Classification of hyperlipidemia ............................................................................. 27

2.6.2 Causes and risk factors of hyperlipidemia ............................................................... 27

2.6.3 Complications of hyperlipidemia ............................................................................ 27

2.6.4 Pathophysiology of hyperlipidemia ........................................................................ 27

2.6.5 Management of hyperlipidemia .............................................................................. 28

2.6.5.1 Non pharmacological management .................................................................. 28

2.6.5.2 Pharmacological therapy.................................................................................. 28

2.6.5.3 Management of hyperlipidemia by natural plants ............................................ 29

Page 10: Computational Approach Towards Exploring the

viii

2.7 Computer aided drug design (CADD( ........................................................................... 30

2.7.1 CADD classification............................................................................................... 31

2.7.2 Computational target fishing .................................................................................. 32

2.7.2.1 Polypharmacology ........................................................................................... 33

2.7.2.2 Drug repurposing ............................................................................................. 34

2.7.2.3 Computational methods for target fishing ........................................................ 35

Chapter Three Methodology ..................................................................................................... 42

3.1 Computational Method .................................................................................................. 42

3.1.1 Building 3D-ligands database ................................................................................. 42

3.1.2 ROCS similarity search .......................................................................................... 43

3.1.3 Docking simulations ............................................................................................... 44

3.1.3.1 Preparation of proteins ..................................................................................... 44

3.1.3.2 Preparation of ligands ...................................................................................... 45

3.1.3.3 Docking ........................................................................................................... 46

Chapter Four Results and discussion ......................................................................................... 47

4.1 Background ................................................................................................................... 47

4.2 Polypharmacologyof S. officinalis ................................................................................. 48

4.2.1 Reported anticancer, antidiabetic and hypolipidemic targets of S. officinalis ........... 48

4.2.2 Targets identified using S. officinalis constituents as queries in RTFA .................... 50

4.2.2.1 Apigenin.......................................................................................................... 50

4.2.2.2 Carnosol .......................................................................................................... 56

4.2.2.3 Cirsimaritin ..................................................................................................... 60

4.2.2.4 Corosolic Acid ................................................................................................ 61

4.2.2.5 Ellagic acid ...................................................................................................... 63

4.2.2.6 Ferruginol ........................................................................................................ 67

4.2.2.7 Genkwanin ....................................................................................................... 70

4.2.2.8 Hispidulin ........................................................................................................ 72

4.2.2.9 Luteolin ........................................................................................................... 75

4.2.2.10 Oleanolic acid ................................................................................................. 77

4.2.2.11 Quercetin ......................................................................................................... 79

Page 11: Computational Approach Towards Exploring the

ix

4.2.2.12 Rutin .............................................................................................................. 82

4.2.2.13 Rosmarinic acid ............................................................................................... 83

4.2.2.14 Ursoli acid ...................................................................................................... 85

4.3 Polypharmacology of Urtica dioica ............................................................................... 86

4.3.1 Reported anticancer antidiabetic and hypolipidemic targets of U. dioica .............. 86

4.3.2 Targets identified using U.dioica constituents as queries in RTFA.......................... 88

4.3.2.1 Caffeoylmalic acid .......................................................................................... 88

4.3.2.2 Chlorogenic acid ............................................................................................. 89

4.3.2.3 Isolariciresinol ................................................................................................. 89

4.3.2.3 Neoolivil ......................................................................................................... 92

4.3.2.5 Secoisolariciresinol ......................................................................................... 95

Chapter Five Conclusion ..................................................................................................... 97

Chapter six Recommendations .............................................................................................. 98

References ............................................................................................................................... 99

Page 12: Computational Approach Towards Exploring the

x

List of Tables

Table 2.1: Ethnomedicinal uses of S. officinalis ......................................................................... 6

Table ‎2.2: Chemical structures of main polyphenols and flavonoides isolated from S. officinalis 7

Table 2.3: Chemical structures of main terpenoids isolated from S. officinalis ........................... 8

Table ‎2.4: Summary of clinical studies on S. officinalis ................................................................ 10

Table 2.5: Ethnomedicinal uses of Urtica dioica L ................................................................... 12

Table 2.6: Structures of chemical constituents of U. dioica ...................................................... 13

Table 2.7: The Anticancer effects of U. dioica ......................................................................... 15

Table 2.8: Various types of anticancer drugs and their examples .............................................. 19

Table 2.9: Anticancerous medicinal plants ............................................................................... 21

Table 2.10: Marketed therapeutic agents for T2DM and respective mechanisms. ..................... 25

Table 2.11: Medicinal plants used in the treatment of T2DM ................................................... 26

Table 2.12: Antihyperlipidemic drugs and their mechanisms of action ..................................... 29

Table ‎2.13: Plants with hypolipidemic activity ......................................................................... 30

Table 3.1: Proteins data obtained from Protein data bank. ........................................................ 44

Table ‎4.1: On-targets and off-targets of S. officinalis ............................................................... 49

Table 4.2: Pharmacological profiling for apigenin using ROCS ............................................... 51

Table ‎4.3: Pharmacological profiling for carnosol using ROCS ............................................... 57

Table 4.4: Pharmacological profiling for cirsimaritin using ROCS ........................................... 60

Table ‎4.5: Pharmacological profiling for corosolic acid using ROCS ....................................... 62

Table 4.6: Pharmacological profiling for ellagic Acid using ROCS .......................................... 63

Table ‎4.7: Pharmacological profiling for ferruginol using ROCS ............................................. 67

Table 4.8: Pharmacological profiling for genkwanin using Rocs……………………………….72

Page 13: Computational Approach Towards Exploring the

xi

Table ‎4.9: Pharmacological profiling for hispidulin using ROcs ............................................... 74

Table 4.10: Pharmacological profiling for luteolin using ROCS ............................................... 75

Table ‎4.11: Pharmacological profiling for oleanolic using ROCS ............................................ 77

Table 4.12: Pharmacological profiling for quercetin using ROCS ............................................ 79

Table ‎4.13: Pharmacological profiling for rutin using ROCS ................................................... 82

Table 4.14: Pharmacological profiling for rosmarinic acid using ROCS ................................... 83

Table ‎4.15: Pharmacological profiling for ursolic acid using ROCS ......................................... 85

Table 4.16: On-targets and off-targets of U. dioica .................................................................. 87

Table ‎4.17: Pharmacological profiling for caffeoylmalic acid using ROCS .............................. 88

Table 4.18: Pharmacological profiling for chlorogenic acid using ROCS ................................. 89

Table ‎4.19: Pharmacological profiling for isolariciresinol using ROCS .................................... 90

Table ‎4.20: Pharmacological profiling for neoolivil using ROcs .............................................. 94

Table 4.21: Pharmacological profiling for secoisolariciresinol using ROCS ............................. 96

Page 14: Computational Approach Towards Exploring the

xii

List of Figures

Figure 2.1: Arial parts of Salvia officinalis L. .......................................................................................... 5

Figure ‎2.2: Parts of U. dioica plant. (a) Whole plant; (b) Flower; (c) Trichomes; (d) Roots; (e) Leaf. .... 11

Figure 2.3: Multiple defects contribute to the development of glucose intolerance in T2DM. ................. 24

Figure ‎2.4: CADD in drug discovery/design pipeline. ........................................................................... 32

Figure 2.5: Comparison between docking and reverse docking .............................................................. 33

Figure ‎2.6: Molecular descriptors based on the ROCS color force field. ................................................ 38

Figure 2.7: Main applications of molecular docking in current drug discovery .......................... 40

Figure ‎3.1: Workflow of ROCS-based target fishing approach (RTFA) ................................................. 42

Figure 3.2: Simple run window display in vRocs user interface after generation of quercetin query. ...... 43

Figure ‎4.1: Mechanism of action for GIP, GLP-1 analogues and DPP4 inhibitors in controlling T2DM . 58

Figure 4.2: A : Detailed view of docked carnosol and the corresponding interacting amino acid within

the binding site of Tank2, B : Detailed view of co-crystallized structure (G9W, PDB

code: 5C5Q) and the corresponding interacting amino acid within the binding site of

Tank2. ............................................................................................................................. 59

Figure ‎4.3: Detailed view of docked cirsimaritin and the corresponding interacting amino acids within

the binding site of PPAR-γ ............................................................................................... 61

Figure 4.4: Mechanism of activation of PKB (AKT), S6K and SGK by PDK1.. .................................... 65

Figure ‎4.5: Insulin signaling.. ................................................................................................................ 66

Figure 4.6: The physiological signal pathways involving PTP1B. .......................................................... 68

Figure ‎4.7: Detailed views of docked feruginol and the corresponding interacting amino acids within

the binding site of PTP1B. ............................................................................................... 69

Figure 4.8: The overview of PI3K/AKT/mTOR signaling pathway.. ...................................................... 71

Figure ‎4.9: A: Detailed view of docked genkwanin and the corresponding interacting amino acid

within the binding site of PIM1, B: Co-crystallized structure (HUL, PDB code: 4XH6)

and the corresponding interacting amino acids within the binding site of PIM1, C:

Overlay view of docked genkwanin within the binding site of EGFR ............................... 73

Page 15: Computational Approach Towards Exploring the

xiii

Figure 4.10: Signaling through MAPK14 cascade and its role in the regulation of cellular functions.. .... 81

Figure ‎4.11: Epidermal growth factor receptor (EGFR) and its downstream signaling proteins. ............. 91

Figure 4.12: A; Detailed view of docked Isolariciresinol and the corresponding interacting amino acid

within the binding site of EGFR, B; Detailed view of co-crystallized structure (1C9 ,

PDB code: 4I23 ) and the corresponding interacting amino acid within the binding site

of EGFR, C; Overlay view of docked Isolariciresinol within the binding site of EGFR ..... 93

Figure 4.13: A: Detailed view of docked Neoolovil and the corresponding interacting amino acid

within the binding site of DPP-4, B: Surface view of docked Neoolovil and the

corresponding interacting amino acid within the binding site of DPP-4. ........................... 95

Page 16: Computational Approach Towards Exploring the

xiv

List of Abbreviations

71β-HSD

17 Beta Hydroxysteriod Dehydrogenase 1

ACAT Acyl Coenzyme-A-Cholesterol Acyl Transferase

ACL Adenosine-Triphosphate Citrate Lyase

ADME/T Absorption, Distribution, Metabolism, Excretion And Toxicity

AKT Protein Kinase B

Aº Angstrom

b GP Brain Glycogen Phosphorylase

Bak Bcl-2 Homologous Antagonist Killer

Bax Bcl-2-Associated Protein X

BCG Bacillus Calmette-Guerin

Bcl2 B-Cell Lymphoma 2

CA Caffeoylmalic Acid

CADD Computer Aided Drug Design

CDK2 Cycline Dependent Kinase 2

CGA Chlorogenic Acid

CK2 Casine Kinase 2

COVID-19 Coronavirus Disease of 2019

Cpd.ID Co-Crystallized ligand Identification

CS Chemical Similarity

CSNAP Chemical Similarity Network Analysis Pull-Down

D Dimension

DAPK1 Death Associated Protein Kinase1

DB Drug Bank

DM Diabetes Mellitus

DMPK Drug Metabolism and Pharmacokinetic

DNA Deoxyribonucleic Acid

DPP-4 Dipeptidyl Peptidase 4

DS Discovery Studio

EA Ellagic Acid

EBV Epstein-Barr Virus

EGFR Epidermal Growth Factor Receptor

EIND Emergency Investigational New Drug

ERK Extracellular Signal-Regulated Kinase

Page 17: Computational Approach Towards Exploring the

xv

ERα Estrogen Receptor alpha

ERβ Estrogen Receptor beta

FDA Food And Drug Administration

FGFR1 Fibroblast Growth Factor Receptor 1

FRED Fast Rigid Exhaustive Protein-Ligand Docking

GIP Glucose-Dependent Insulin-Tropic Polypeptide

GLO-1 Glyoxalase

GLP-1 Glucagon Like Peptide 1

GLUT-4 Glucose Transporter 4

GP Glycogen Phosphorylase

GPR G-Protein Coupled Receptor

HbA1c Glycated Haemoglobin

HER2 Human Epidermal Growth Factor Receptor 2

HLD High Density Lipoprotein

HMG-CoA Hydroxy-3-Methylglutaryl Coenzyme A

HPVs Human Papilloma Viruses

HSP90-α Heat Shock Protein 90 alpha

IARC International Agency for Research on Cancer

IC50 Half-Maximal Inhibitory Concentration

IDDM Insulin Dependent Diabetes Mellitus

IDF International Diabetes Federation

ITC Isothermal Titration Calorimetry

Ki Inhibition Constant

KSHV Kaposi‘s Sarcoma-Associated Herpes Virus

l GP Liver Glycogen Phosphorylase

LBDD Ligand Based Drug Design

LDL Low Density Lipoprotein

LHRH Luteinizing Hormone Releasing Hormone

LPL Lipoprotein Lipase

m GP Muscle Glycogen Phosphorylase

MAPK14 Mitogen Activated Protein Kinase 14

Mcl-1 Myeloid Leukemia Cell Differentiation Protein 1

MMp Metalloproteinase

MOH Ministry of Health

MTP Microsomal Triglyceride Transfer Protein

mTOR Mammalian Target of Rapamycin

Page 18: Computational Approach Towards Exploring the

xvi

NCI National Cancer Institution

NF-kB Nuclear Transcription Factor-Kappa B

NIDDM Non–Insulin Dependent Diabetes Mellitus

NMR Nuclear Magnetic Resonance

NPs Natural Products

OA Oleanolic Acid

PARP Poly Adenosine-Diphosphate (ADP)-Ribose Polymerase

PCSK9 Proprotein Convertase Subtilisin/ Kexin type 9

PDB Protein Data Bank

PDK1 Phosphoinositide Dependent Protein Kinase 1

Pgp P-Glycoprotein

PI3Kγ Phosphatidylinositol 3-Kinase gamma

PIK3CG Phosphatidylinositol-4,5-Bisphosphate3-Kinase Catalytic Subunit gamma

PIK3Cα Phosphatidylinositol-4,5-Bisphosphate3-Kinase Catalytic Subunit alpha

PIM1 Proviral Integration Site for Moloney Murine Leukaemia Virus 1

PIN1 Peptidyl-prolyl cis/trans Isomerase Never In Mitosis A (NIMA)-

Interacting 1

PIP3 Phosphatidylinositol (3,4,5)-Triphosphate

PKACα Protein Kinase A Catalytic Subunit alpha

PMoH Palestinian Ministry Of Health

PPAR-α Peroxisome Proliferator Activated Receptor alpha

PPAR-γ Peroxisome Proliferator Activated Receptor gamma

PTEN Phosphatase and Tensin Homolog

PTP1B Protein Tyrosine Phosphatase 1B

QSAR Quantitative Structure Activity Relationship

RA Rosmarinic Acid

RAF Rapidly Accelerated Fibrosarcoma Kinase

Ras Rat Sarcoma Gene

RCS Reactive Chloride Species

Ref References

RNS Reactive Nitrogen Species

ROCS Rapid Overlay of Chemical Structures Software

ROS Reactive Oxygen Species

RPTP-γ Receptor Type Protein Tyrosine Phosphatase gamma

RS Reactive Species

RSS Reactive Sulfur Species

RTFA ROCS Based Target Fishing Approach

Page 19: Computational Approach Towards Exploring the

xvii

RTK Receptor Tyrosine Kinase

S. Salvia

SBDD Structure Based Drug Design

SGLT2 Sodium Glucose Co-Transporter 2

SIRT-1 Silent Information Regulator Proteins-1

T1DM Type 1 Diabetes Mellitus

T2DM Type 2 Diabetes Mellitus

Tank2 Tankyrase 2

TG Triglycerides

U. Urtica

UA Ursolic Acid

VEGF Vascular Endothelial Growth Factor

VEGFR Vascular Endothelial Growth Factor Receptor

VLDL Very Low Density Lipoprotein

VS Virtual Screening

WHO

World Health Organisation

Page 20: Computational Approach Towards Exploring the

4

1 Chapter One

Introduction

1.1 Background

Natural products and traditional medicines are of great importance. They have been recognized

for many years as a source of therapeutic agents and have shown beneficial uses. Discovery of a

new drug usually faces challenges in the form of time and money investment. On the other hand,

traditional medicine is affordable, of low level of side effects, easily accessible, and culturally

familiar (Katiyar, et al., 2012).

The world is decorated with medicinal herbs, which is a rich wealth of endurance. Every plant is

identified by its own different therapeutic properties due to the bioactive molecule. Herbal

medicine is based on traditional medicines which have their own importance and basic

philosophy. So exploration of the chemical constituents of the plants and their pharmacological

screening may provide us the basis for developing a lead molecule through herbal drug discovery

(Kar, 2006). Thus, there is a need of investigating the various bioactive fractions as well as

evaluating of pharmacological activity of herbal drugs for achieving the dreams of herbal drug

discovery.

In our research we have chosen two of these herbs to investigate their role in treatment

of serious diseases such as cancer, diabetes and hyperlipidemia. The two herbs are

Salvia officinalis and Urtica dioica which are widely used as a medicine due to their

many pharmacological and clinical effects.

Salvia officinalis (S. officinalis) is a plant from the family of Labiatae/ Lamiaceae. A wide range

of phytochemicals are well identified in S. officinalis include alkaloids, flavonoids, glycosidic

derivatives, phenolic compounds, steroids, terpenes/ terpenoids and waxes. Studies have

revealed many pharmacological activities of Salvia officinalis include anticancer, hypoglycemic,

hypolipidemic, anti-inflammatory, antinociceptive, antioxidant, antimicrobial, antimutagenic,

and antidemential effects (Badiee, et al., 2012).

Urtica dioica (U. dioica) belongs to Urticaceae family, is a perennial herb commonly known as

‗stinging nettle‘. Phytochemical studies revealed the presence of many valuable chemical

compounds such as alkaloids, tannins, flavonoids, steroids, terpenes and polyphenols. Urtica

dioica has been reported to have various pharmacological activities like antibacterial,

Page 21: Computational Approach Towards Exploring the

2

antioxidant, analgesic, anti-inflammatory, antiviral, im munomodulatory, hepatoprotective, and

anticancer effects (Joshi, et al., 2014).

Identifying the complete target spectrum of bioactive compounds is not only a critical step in the

understanding of the mechanism of action, but also is of particular interest for rationally

designing more effective and less toxic drugs, predict the adverse effects of a compound, or drug

repurposing (Cereto-Massague, et al., 2015).

In our research, we will use in-silico target fishing approach in order to explore potential new

targets for Salvia officinalis and Urtica dioica natural compounds focusing on anticancer,

antidiabetic and antihyperlipidemic targets.

1.2 Problem statement

On the basis of the available literature evidence, S. officinalis shows many pharmacological

effects. The possible therapeutic applications for these effects need to be elucidated (Ghorbani &

Esmaeilizadeh, 2017). Also, further investigations are necessary to understand the exact

molecular mechanisms responsible for the effects of S. officinalis , especially anticancer,

antidiabetic and antihyperlipidemic activities.

Urtica dioica is used for the treatment of various diseases due to its remarkable power of

healing. This plant has got the place among the top ranked evidence based herbal medicines

(Sepide, 2016). In folk medicine U. dioica was used for the treatment of arthritis and it showed

the presence of antiasthmatic, antidandruff, astringent, depurative, diuretic, galactogogue,

haemostatic and hypoglycaemic activities in preclinical experiments. Exploration of the exact

mechanisms of actions of U. dioica and its phytoconstituents are required to support its

traditional uses (Mueen & Parasuraman, 2014), especially anticancer, antidiabetic and

antihyperlipidemic activities.

1.3 Aim

To explore the polypharmacology of Urtica dioica and Salvia officinalis focusing on anticancer,

antidiabetic and antihyperlipidemic targets.

Page 22: Computational Approach Towards Exploring the

2

1.4 Objectives

To identify new potential anticancer, antidiabetic and antihyperlipidemic targets for the

natural U. dioica components.

To identify new potential anticancer, antidiabetic and antihyperlipidemic targets for the

natural S officinalis components.

To emphasize the rule of the components of U. dioica and S. officinais as anticancer,

antidiabetic and antihyperlipidemic agents.

To open new space for further researches to evaluate the natural compounds of U. dioica

and S. officinalis as lead compounds for new anticancer, antidiabetic and antihyperlipidi-

mic targets.

1.5 Justification of the Study

Herbal medicines are very important to cure the various ailments of human. Demands of the

herbal medicines are increasing in both developed and under developed countries due to growing

recognition of natural plants being lesser of side effect, easily available in the surrounding

geographical area with low cost.

Traditional medicine using herbal drugs exists in every part of the world. U. dioica and S.

officinalis are widely distributed in Palestine and have the advantage of being available for

patients in the surrounding places with low cost and less side effects.

In addition, cancer, diabetes mellitus (DM) and hyperlipidemia are serious problems affecting

humankind, with significantly increasing number of cases and deaths. At the same time, the

prevailing treatments are not effective enough and cause adverse side effects, which justifies the

need to discover other effective and safe agents.

Moreover, the use of in-silico target fishing approach to explore the polypharmacology of

endogenous medicinal plants is the first time to be applied in Palestine. This encourage and

attract us to investigate these aspects to well distributed and known medicinal herbs in our

country, U. dioica and S. officinalis, aiming to identify new potential targets that could help in

designing new therapeutic agents for the treatment of serious diseases such as cancer, diabetes

mellitus and hyperlipidemia.

Page 23: Computational Approach Towards Exploring the

1

2 Chapter Two

Literature Review

2.1 Drug discovery and importance of medicinal plants

Drug discovery using natural products (NPs) is a challenging task for designing new leads. It

involves a wide range of scientific disciplines, including biology, chemistry and pharmacology.

Research in drug discovery needs to develop robust and viable lead molecules, which step

forward from a screening hit to a drug candidate through structural elucidation and

identification. The development of new technologies has revolutionized the screening of natural

products in discovering new drugs. Utilizing these technologies gives an opportunity to perform

research in screening new molecules using a software and database to establish natural products

as a major source for drug discovery (Koparde, et al., 2019).

Natural products are one of the main sources of drug discovery. According to the data from

Newman, most new FDA-approved drugs between 1981 and 2014 were derived from NP

structures (Newman & Cargg, 2016).

Natural constituents are widely distributed in various natural sources, including plants,

microorganisms and invertebrates. Plant-derived molecules continue to make up a large portion

of the pharmaceuticals in the clinic. The most famous example to date is probably the synthesis

of the anti-inflammatory agent acetylsalicylic acid (aspirin), derived from salicin and isolated

from the bark of the willow tree Salix alba L . Other examples are morphine, codeine, digitoxin,

quinine and the antitumor agents paclitaxel, vincristine and vinblastine, and a long list of other

drugs. In addition, the production of antibiotics by microorganisms was one of the biggest

breakthroughs in the history of drug discovery in the twentieth century (Ramos, et al., 2018).

To better understand the huge impact of NPs, for example, on cancer pharmaceuticals, it is worth

mentioning that out of 155 small molecules used as chemotherapeutics, 73 are directly NPs and

another 40 are derivatives or synthetic NP mimetics (Newman & Cragg, 2007). Furthermore,

current research trends in the field suggest an optimistic future for NPs in cancer prevention and

new therapeutics drug discovery.

Because of the complex chemistry generated by centuries of evolution of NPs, more success is

expected in drug discovery with NPs than with synthetic molecules. However, that complexity of

the natural molecules requires a coordinated effort from the interaction of multidisciplinary

Page 24: Computational Approach Towards Exploring the

5

research areas with new and more sophisticated analytical and technical expertise in order to

extract, isolate, identify and turn them into promising leads (Ramos, et al., 2018).

Since only a small amount of the world‘s biodiversity has been evaluated for potential biological

activity, many more useful natural lead compounds await discovery, the challenge being how to

access this natural chemical diversity (Ramos, et al., 2018).

2.2 Sage

Salvia officinalis L. is a plant from the mint family Lamiaceae, subfamily Nepetoideae, tribe

Mentheae, and genus Salvia. Salvia is the largest genus of the Lamiaceae family, containing

around 1000 species. It is known as garden sage or common sage as seen in figure 2.1. It is an

aromatic perennial woody sub-shrub native to the mediterranean area region and widely

distributed over the hillsides and shores of southern Europe. Today's, it has been naturalized

throughout the world. It is cultivated throughout Europe and the USA, including Spain, Italy,

Yugoslavia, Greece, Albania, Argentina, Germany, France, Malta, Turkey, England and Canada

(Sharma & Schaefer, 2019).

Figure ‎2.1: Arial parts of Salvia officinalis L. (Ghorbani & Esmaeilizadeh, 2017)

Sage is a plant of both tropics and temperate region, grown for food, home remedies, and

commercial pharmaceuticals. It is a multipurpose plant, on which several research studies were

Page 25: Computational Approach Towards Exploring the

6

reported supporting its traditional utilization, biological effects, and mode of action (Sharma &

Schaefer, 2019).

2.2.1 Ethnomedicinal uses of S. officinalis

Historically, sage is known as the ―Salvation Plant‖, originating from the old Latin word

―salvarem‖, which means save or cure. It has been used to reduce perspiration, as a gargle for

sore throat, to improve regularity of a menstrual cycle and to reduce hot flashes in menopause, to

fight gastroenteritis and other infections, to improve lipid status and liver function in general, to

improve appetite and digestion, and to improve mental capacity (Jakovljevicet, et al., 2019).

Traditional medicinal uses of S. officinalis documented by different communities and regions of

the world are presented in table 2.1 (Sharma & Schaefer, 2019).

Table ‎2.1: Ethnomedicinal uses of S. officinalis (Sharma & Schaefer, 2019)

Region Traditional use

Europe and the

Mediterranean region

Sage leaf tea : relieve stress, indigestion, bloating, heartburn, acidity, sore throat,

and sunburn

Leaf extract : treat excessive sweating, bronchitis and Alzheimer‘s disease

Jordan Leaf extract : used as antibacterial, anti-inflammatory and antiseptic

South east Asia and India Fresh leaves and decoction: antispasmodic, refreshing tea, hypotensive and to treat

respiratory disorders

Latin America Sage tea and essential oil : treat convulsion, nerve related disorders and high blood

pressure

Turkey, Serbia, and Iran

Flower, leaf and stem extracts : as antiseptic in wounds, pharyngitis, mouth ulcers

and consumed internally for dysmenorrhea.

Leaf tea is : stimulant, digestive, sedative and analgesic

Brazil

Hydroalcoholic tincture and leaf tea : to relieve stress, inflammation, prevent

excessive bleeding and as a mouth freshener. Commonly used to increase memory

power.

Greeks and Romans

leaves : as a spice and appetite stimulant for easy digestion of fatty foods

Leaf tea : for ulcers, sore throats, and laryngitis

Leaf decoction : enhance memory and brain power

Valencia region of Spain

leaf decoction : as an appetizer and hypotensive .

Hot tea : for detoxification, cold, and throat infections Dry leaves : as a spice and essential oil extraction

2.2.2 Phytochemical constituents of S. officinalis

The major phytochemicals in flowers, leaves, and stem of S. officinalis are well identified. A

wide range of constituents include alkaloids, carbohydrate, fatty acids, glycosidic derivatives

(e.g., cardiac glycosides, flavonoid glycosides, saponins), phenolic compounds (e.g., coumarins,

flavonoids, tannins), polyacetylenes, steroids, terpenes/terpenoids (e.g., monoterpenoids,

Page 26: Computational Approach Towards Exploring the

7

diterpenoids, triterpenoids, sesquiterpenoids) , and waxes are found in S. officinalis (Ghorbani &

Esmaeilizadeh, 2017).

Several polyphenolic compounds like chlorogenic acid, ellagic acid, epicatecin, epigallocatechin

gallate and rosmarinic acid, flavonoids like quercetin, and rutin as well as several volatile

components such as borneol, cineole, camphor, elemene, α-pinene, and thujone have been

identified in infusion prepared from S. officinalis (Ghorbani & Esmaeilizadeh, 2017).

The chemical structures of the main polyphenols and flavonoids as well as terpenoids isolated

from S. officinalis are shown in table 2.2 and table 2.3, respecively (Ghorbani & Esmaeilizadeh,

2017).

Table ‎2.2: Chemical structures of main polyphenols and flavonoides isolated from S. officinalis

O

OH

OOH

HO

O

OH

OOH

O

O

O

OH

OOH

O

Apigenin (C15H10O5) Circimaritin (C17H14O6) Genkwanin (C16H12O5)

O

OH

OOH

HO

OH

OHO

OH O

OH

OH

OH

Hispidulin (C16H12O6) Luteolin (C15H10O6) Quercetin (C15H10O7)

OHO

OH O

OH

OH

O

O

OO

OH

OH

OH

OH

OH

HO

HO O

O

OH

OHO

OH

OH

O

O

O

OH

OH

O

HO

HO

Rutin (C27H30O16) Rosmarinic acid (C18H16O8) Ellagic acid (C14H6O8)

The highest yield of total phenolics and flavonoids in sage extracts was obtained using 40%

aqueous ethanol solution which confirms that water ethanol solvents are probably the most

suitable for extraction of phenolic compounds and flavonoids from the sage due to the different

polarity of the bioactive constituents, and the acceptability of this solvent system for human

consumption (Osmic, et al., 2019).

The most common monoterpenes include: α- and β-thujone, 1, 8-cineole, and camphor. The most

common diterpenes include: carnosic acid, carnosol, rosmadial, and manool. Triterpenes include

O

OH

OOH

HO

O

Page 27: Computational Approach Towards Exploring the

8

oleanolic acid, corosolic acid and ursolic acid. In addition, sesquiterpenes as α-humulene and

viridiflorol are also present in sage extracts (Jakovljevic, et al., 2019).

Table ‎2.3: Chemical structures of main terpenoids isolated from S. officinalis

OH

H

O

α-Pinene (C10H16) Borneol (C10H18O) Camphor (C10H16O)

OH

H

O

O

HO

H

H

O

Carnosol (C20H26O4) Caryophyllene (C15H24)

Cineole (C10H18O)

COOHH

HO

H

H

HO

Corosolic acid (C30H48O4) Cymene (C10H14) Elemene (C15H24)

HO

H

Ferruginol (C20H30O) Limonene (C10H16) Myrcene (C10H16)

COOH

HO

H

H

H

O

H

HO

H

HOH

OH

Oleanolic acid (C30H48O3) Thujone (C10H16O) Ursolic acid (C30H48O3)

2.2.3 Pharmacological activities of S. officinalis

2.2.3.1 Anticancer effects

Extracts of S. officinalis showed proapoptotic and growth inhibitory effects on cell lines of breast

cancer, cervix adenocarcinoma, colorectal cancer, insulinoma, laryngeal carcinoma, lung

carcinoma, melanoma, and oral cavity squamous cell carcinoma. In addition to antiapoptotic

Page 28: Computational Approach Towards Exploring the

9

action, S..officinalis has antiproliferative, antimigratory and antiangiogenic effects (Kontogianni,

et al., 2013).

These effects may be related to the presence of several cytotoxic and anticancer compounds in S.

officinalis as ursolic and rosmarinic acids. Ursolic acid inhibits angiogenesis and invasion of

melanoma cells (Jedinak, et al., 2006). Rosmarinic acid inhibits the growth of various human

cancer cells including breast adenocarcinoma, colon, chronic myeloid leukemia, prostate,

hepatocellular and small cell lung carcinoma (Yesil-Celiktas, et al., 2010).

2.2.3.2 Antidiabetic effects

Recent pharmacological investigations demonstrated that different extracts of aerial parts of S.

officinalis are able to decrease blood glucose in normal and diabetic conditions. The mechanisms

suggested for hypoglycemic effect of S. officinalis include an inhibition of hepatocyte

gluconeogenesis and decrease of insulin resistance (Christensen, et al., 2010, Ghorbani &

Esmaeilizadeh, 2017).

2.2.3.3 Antihyperlipidemic effects

In clinical trials, extract of S. officinalis leaf could lower the blood levels of triglyceride, total

cholesterol, low density lipoproteins (LDL), very low density lipoproteins (VLDL) and two hour

postprandial glucose in patients with hyperlipidemia and diabetes. The beneficial properties of S.

officinalis tea consumption on serum lipid profile have been also reported on nondiabetic healthy

volunteers (Kianbakht & Dabaghian, 2013). This activity may be related to flavonoids present in

the plant such as rosmarinic acid and rutin (Govindaraj & Sorimuthu, 2015).

2.2.3.4 Other effects

Several studies have established that extracts of S. officinalis possess various pharmacological

effects, including antioxidant (Ghorbani & Esmaeilizadeh, 2017), anti-inflammatory and

antinociceptive properties (Azevedo, et al., 2013), antimicrobial effects (Badiee, et al., 2012) as

well as cognitive and memory-enhancing effects (Miroddi, et al., 2014).

2.2.4 Clinical studies

Clinical studies on pharmacological properties of S. officinalis are summarized in table 2.4

(Ghorbani & Esmaeilizadeh, 2017).

Page 29: Computational Approach Towards Exploring the

41

Table ‎2.4: Summary of clinical studies on S. officinalis (Ghorbani & Esmaeilizadeh, 2017).

Category Study design Subjects Dosage Effects

Effects on

memory and

cognitive

functions

Randomized

placebo controlled

trial

Patients with

Alzheimer's

Disease

60 drops/day of

alcoholic extract

For 16 weeks

Improvement of cognitive

Functions

Randomized

placebo controlled

trial

Healthy young

participants

300-600 mg

encapsulated

dried leaf

Improvement of mood and

cognitive functions after

single Dose

Randomized

placebo controlled trial

Healthy old

participants

165-132 mg of ethan-

olic extract was administrated 1,2,4

and 6 hours before

assessment

Improvement of memory and

Attention

Randomized

controlled trial

Healthy adults

participants

5 drops of essential

oil were placed into

the testing cubicle

Improvement of prospective

memory and cognitive

performance

Effects on

pain

Randomized

controlled trial

Patient with

pharyngitis

15% spray containing

140 ml of the plant

extract per dose

Reduction of the throat pain

Intensity

Effects on

glucose

and lipids

Randomized

placebo controlled

trial

Patients

diagnosed with

primary

hyperlipidemia

500 mg encapsulated

hydroalcoholic

extract every 8 h for

2 months

⇊ of the blood levels of total

cholesterol, triglyceride, LDL

and VLDL;

Increase of HDL Level

Randomized

placebo controlled

trial

Hyperlipidemic

type 2 diabetic

patients

150 mg sage extract

3 times a day for

3 months

⇊ of the blood levels of total

cholesterol, glucose, HbA1c,

triglyceride, and LDL; Increase of HDL level

Randomized

placebo controlled

trial

Type 2 diabetic

patients

300 mL of sage

tea twice daily

for 4 weeks

No effect on fasting glucose,

HbA1c, triglyceride, LDL and

HDL

A pilot study (non-randomized trial)

Healthy female

volunteers

300 mL of sage tea twice daily

for 4 weeks

⇊ of total cholesterol and LDL;

No effect on fasting glucose;

Increase of HDL level

HbA1c: Glycated haemoglobin, HDL: High density lipoprotein, LDL: Low density lipoprotein, VLDL: Very

low density lipoprotein, ⇊: Reduction

2.3 Stinging Nettle

Nettle, or stinging nettle, is a perennial plant that is widely distributed throughout the temperate

and tropical areas around the world. It grows from two to four meters high and produces pointed

leaves and white to yellowish flowers and it belongs to the family of Urticaceaea and to the

genus of Urtica as seen in figure 2.2 (Joshi, et al., 2014).

The genus name Urtica comes from the Latin verb urere, meaning ‗to burn,‘ because of these

stinging hairs with a tuft (a small cluster of elongated flexible outgrowths attached or close

together at the base and free at the opposite ends) of hair at the apex. Leaves and stems contain

abundant non-stinging hairs, with touch sensitive tips, needles that will inject chemicals

including serotonin, histamine,

Page 30: Computational Approach Towards Exploring the

44

acetylcholine, leukotrienes and possibly formic acid into the skin. The irritant compounds

provoke pain, wheals or a stinging sensation (Fu, et al., 2006).

The species name dioica means ‗two houses‘ because the plant usually contains either male or

female flowers. In the last few years, Urtica dioica L., has been accepted as a healing plant

because of its considerable effects on human health in many countries all over the world (Joshi,

et al., 2014).

Figure ‎2.2: Parts of U. dioica plant. (a) Whole plant; (b) Flower; (c) Trichomes; (d) Roots; (e) Leaf (Joshi, et

al., 2014).

2.3.1 Ethnomedicinal uses of U. dioica

U. dioica is widely used by the traditional medicinal practitioners for curing various diseases

such as nephritis, haematuria, jaundice, menorrhagia, arthritis and rheumatism. The plant also

has been used as food, fiber, paint, manure and cosmetics (Joshi, et al., 2014). The

ethnomedicinal uses of stinging nettle in various countries are shown in table 2.5.

2.3.2 Phytochemical constituents of U. dioica

The phytochemical composition investigation on U. dioica revealed that it contains different

compounds, incuding alkaloids, terpenoids, flavonoids, phenolic acids, sterols, fatty acids,

Page 31: Computational Approach Towards Exploring the

42

polysaccharides and lignans (Esposito, et al., 2019). The categorizations of these compounds,

their molecular formulas and chemical structures are listed in table 2.6 (Ibrahim, et al., 2018).

Table ‎2.5: Ethnomedicinal uses of Urtica dioica L (Joshi, et al., 2014).

Region Ethnomedicinal uses

Brazil

Asthma, bronchitis, cough, bleeding, diabetes, diarrhea, dysentery, fever, liver support, lung

problems, menstrual disorders, pneumonia, skin disorders, ulcers, urinary problems, and to

increase perspiration

Cuba Burns, flu, hemorrhoids, urinary insufficiency and to treat wounds

Germany Arthritis, inflammation, prostate diseases, rheumatism, urinary insufficiency and urinary tract

disorders

Greece Asthma, inflammation, laxative, pleurisy, spleen disorders and urinary insufficiency

India Eczema, nosebleeds, skin eruptions and uterine haemorrhages

America Allergies, arthritis, bleeding, hair loss, hypertension, inflammation, prostatitis, rhinitis, sinusitis,

urinary insufficiency and wounds

In particular, it was observed that all the parts (roots, stalk and leaves) of U. dioica are a rich

source of phenols and polyphenols and that their content is higher in wild plants than in

domesticated plants. The polyphenol profile seems to be strongly dependent on the parts of the

plant investigated, but also on the harvest site and season (Esposito, et al., 2019).

Root samples from mediterranean cultivar were reported to contain phenol compounds, such as

ferulic acid and polyphenols such as naringin, ellagic acid, myricetin and rutin. The roots also

contained lignans (secoisolariciresinol, 9,9΄-bisacetyl-neo-olivil and their glucosides), phytoste-

rols (e.g., β-sitosterol), coumarins (e.g., scopoletin), simple phenols (e.g., p-hydroxybenz-

aldehyde), triterpenoic acids (e.g., oleanolic acic and ursolic acid) and monoterpendiols (e.g,

carvacrol, carvone) (Gul, et al., 2012; Esposito, et al., 2019).

U. dioica leaves are also constituted by flavonoid glycosides, mainly rutinosyl flavonols.

Chlorogenic acid and caffeoyl malic acid represented approximately 76.5% of total phenolic

compounds, whereas rutin was the most abundant flavonol derivative. Isorhamnetin-3-O-

rutinoside was found, together with rutin, quercetin-3-O-glucoside and kaempferol-3-O-

glucoside in methanolic extracts of U. dioica leaves and stalks (Otles & Yalcin, 2012).

Among lipid secondary metabolites, carotenoids were detected in the leaves and their total

content was estimated equal to 29.6 mg/100 g dry weight (Esposito, et al., 2019).

Page 32: Computational Approach Towards Exploring the

42

The leaves are rich in vitamins B, C and K as will as minerals such as calcium, iron, magnesium,

phosphorus, potassium and sodium (Wetherilt, 1992; Gul., et al., 2012).

Table ‎2.6: Structures of chemical constituents of U. dioica

O

OH

HO

HO

O

OH

O

O

O

OHHO

HO

O O

O

HO OH

HO

HO

Neoolivil (C20H24O7) Neoolivil; 4-O-b-d-Glucopyranoside (C26H34O12)

O

HO

O

OH

OH

OH

HO

O

OH

O

OH

HO

Isolariciresinol (C20H24O6) Secoisolariciresinol (C20H26O6)

Lignanes

O

O HO

OHHO

O

OH

HO

HO

O

OH

OH

O

O

O

HO

HO

Chlorogenic acid (C16H18O9) Caffeoylmalic acid (C13H12O8)

H

H

O

HO

OH

OH

OH

O

H

H

HO

O

Caffeic acid (C9H8O4) Ferulic acid (C10H10O4)

Phenolic acids

OHO

OH O

OH

OH

OH

O

HO

HO

O OH

OH

OH

OH

Quercetin (C15H10O7) Myricetin (C15H10O8)

Page 33: Computational Approach Towards Exploring the

41

O

OO

HO

OH

OH

OHHO

HO

O

OH

OH

OHO

OH O

OH

OH

O

O

OO

OH

OH

OH

OH

OH

HO

Isoquercitrin (C21H20O12) Rutin (C27H30O16)

Flavonoides

OH OH HO

O

O O

9,10 Pinanediol (C10H18O2) Diocanol (C16H22O4)

COOH

HO

H

H

H

HO

H

HOH

OH

Oleanolic acid (C30H48O3) Ursolic acid (C30H48O3)

H

H

H

HO

β-Amyrin (C30H50O) Carvacrol (C10H14O)

Terpenoides

2.3.3 Pharmacological activities of U. dioica

2.3.3.1 Antidiabetic effects

The aqueous extract of U. dioica has shown a significant glucose lowering effect against alloxan

induced diabetes in rats (Bnouham, et al., 2003). The fructose induced insulin resistance in male

rats has been shown to decrease serum glucose level on administration of hydroalcoholic leaf

extract (Ahangarpour, et al., 2012). The cold methanolic extract of leaves has also shown

significant antihyperglycemic effect in alloxan induced diabetes (Al-Wasfi, et al.,2012).

OH

Page 34: Computational Approach Towards Exploring the

45

2.3.3.2 Antihyperlipidemic effects

The plant has very potent antihyperlipidemic activity as it lowers the levels of lipids and

lipoproteins in blood. The aqueous extract of the plant at a dose of 150 mg/kg given for 30 days

to rats fed on normal or high-fat diet, improved the blood lipid profile (Daher, et al., 2006). The

ethanolic extract of the plant at a dose of 100 and 300 mg/kg has shown significant reduction in

the level of total cholesterol and LDL level in hypercholesterolemic rats (Avci, et al., 2006).

2.3.3.3 Anticancer effects

Various studies have recently demonstrated the cytotoxic and anticancer properties of U. dioica,

in particular, against colon, gastric, lung, prostate and breast cancers. Table 2.7 summarizes the

main findings regarding the anticancer properties of U. dioica with the plant extracts and parts

used (specifying collection sites), the cancer type or animal models tested and the effects.

(Esposito, et al., 2019).

Table ‎2.7: The Anticancer effects of U. dioica (Esposito, et al., 2019)

Used parts (site) Extracts Cancer Type Effects

roots (Iran) Ethanolic extract Human colon and gastric cancer ↓Proliferation, ↑Apoptosis

Aerial parts (Iran) Dichloromethane

extract human colon cancer ↓Proliferation, ↑Apoptosis

Leaves (Italy) Methanolic extract human lung cancer ↓Proliferation , ↑Apoptosis

↑caspase3

leaves (Iran) Aqueous extract prostate cancer ↓Proliferation, ↑Apoptosis

Root (Germany) Methanolic extract Human prostate cancer ↓Proliferation

Leaves (Iran) Dichloromethane

extract human breast cancer

↓Proliferation, ↑Apoptosis ,

↑caspase 3 and 9

Leaves (Germany) Dichloromethane

extract Mouse model of breast cancer

↓Metastasis , ↑Apoptosis ,

↑caspase 3 and 9

leaves and stems

(Jordan) Ethanol extract Human breast cancer ↓Proliferation

2.3.3.4 Other effects

Several studies have established that extracts of U. dioica possess various pharmacological

effects including analgesic and anti-inflammatory (Hajhashemi & Klooshani, 2013), antioxidant

(Gulcin, et al., 2004), antimicrobial (Turker, et al., 2008), immunomodulatory (Akbay, et al.,

2003), diuretic (Dizaye, et al, 2013) antiviral (Balzarini, et al., 2005), hepatoprotective and

anthelmintic (Kataki, et al., 2012).

Page 35: Computational Approach Towards Exploring the

46

2.4 Cancer

Cancer is a group of diseases characterized by the uncontrolled growth and spread of abnormal

cells. If the spread is not controlled, it can result in death. Other terms used are malignant tumors

and neoplasms (Costa, 2020)

Cancer is a major public health problem and the second leading cause of death worldwide while

the long-term prognosis is still unfavorable. The global cancer burden is estimated to have risen

to 18.1 million new cases and 9.6 million deaths in 2018. 1 in 5 men and 1 in 6 women

worldwide develop cancer during their lifetime, and one in 8 men and one in 11 women die from

the disease (International Agency for Research on Cancer, IARC, 2018).

Lung, prostate, colorectal, stomach and liver cancer are the most common types of cancer in

men, while breast, colorectal, lung, cervix and thyroid cancer are the most common among

women. Cancers of the lung, female breast, and colorectal are the top three cancer types in terms

of incidence, and are ranked within the top five in terms of mortality (first, fifth, and second,

respectively). Together, these three cancer types are responsible for one third of the cancer

incidence and mortality burden worldwide (IARC, 2018).

In 2015, Palestinian Ministry of Health official statistics revealed that the rate of cancer patients

in Palestine reached 83.8 per one 100 thousand persons, 83.9 cases per 100 thousand persons in

Gaza and 83.8 cases per 100 thousand persons in the West Bank. 52.5% of the new cancer cases

are females and 47.4% are males. Breast cancer ranks first since it constitutes 17.8% of all

cancer cases. Breast cancer also came first for cancers that affect women in Palestine, which

reached 33.7% with a rate of 33.1 new cases per 100 thousand females in Palestine annually,

Colon Cancer comes second regarding cancer cases with the rate of 9.4%. Lung cancer comes

third with the rate of 8.7%, but it its ranked first for cancers that affect males (PMoH, 2016).

2.4.1 Causes & risk factors of cancer

It is impossible to know why some people get cancer unlike others who don't, but there are many

risk factors that increase the possibility of some people to develop cancer without others. The

origin and advancement of cancer depend on many factors inside the cell as well as factors

external to the cells. The inside cell factors include inherited genetics (as Tumor suppressor

genes, oncogenes and DNA repair genes), mutations, immune conditions, hormones and ageing

while smoking, alcohol use ,unhealthy diet ,physical inactivity, chemicals, radiations, bacterial

infections as Helicobacter pylori and viral Infections as Human Papilloma Viruses (HPVs),

Page 36: Computational Approach Towards Exploring the

47

Epstein-Barr virus (EBV) and Kaposi‘s sarcoma-associated herpes virus (KSHV) represent the

external factors (NCI, 2015b).

These entire elements act together to cause abnormal cell behavior and uncontrolled

proliferation. Cancer develops as a result of changes to genes that control cells functions

specially growth and division. These genetic changes (mutations) might be inherited or acquired

during one's lifetime as chemical and radiation exposure (NCI, 2015a). Some genes are

completely linked to develop cancer which fall into 3 groups:

Tumor suppressor genes: are defensive genes such as p53. Normally, these genes slow down

cell growth, repair damaged DNA and control cells death (apoptosis). If a tumor suppressor gene

is missed or damaged, cells divide in uncontrolled manner and may eventually form a tumor.

More than 50% of all cancers involve mutated p53 gene (Rivlin, et al., 2011; NCI, 2015b).

Oncogenes: genes promote normal cell to turn into a cancerous one. The most common

oncogenes are Ras (Rat sarcoma gene) family and HER2 (Human epidermal growth factor

receptor 2). Ras is responsible for translating proteins involved in cell signaling pathways, cell

division, and cell survival (NCI, 2015b).

DNA repair genes: genes responsible for correcting DNA damage, so default gene won‘t

correct the damage of DNA and may lead to develop cancer (NCI, 2015b).

2.4.2 Oxidative stress and cancer

Eukaryotic cells generate energy through aerobic respiration process and free radicals (reactive

species RS) are produced as a result. Free radicals are classified into four groups, reactive

oxygen species (ROS), reactive nitrogen species (RNS), reactive sulfur species (RSS) and

reactive chloride species (RCS). ROS is the most abundant product that includes superoxide

anion (O2-), hydrogen peroxide (H2O2), hydroxyl radical (OH

-) singlet oxygen (

1O2) and ozone

(O3) (Sosa et al., 2013). ROS can serve as anticancer (e.g. through promoting cell cycle stasis,

apoptosis, senescence, necrosis and inhibiting angiogenesis) or as procancer (through promoting

angiogenesis, proliferation, metastasis, invasiveness and suppressing apoptosis). Other types of

RS have the same two faces (Halliwell, 2007). What determines which face will show up is the

equilibrium status between ROS oxidants and ROS scavengers, low concentration of ROS is

beneficial, unlike harmful high concentration of ROS (Reuter, 2010). This ROS-high

concentration is called oxidative stress. Oxidative stress is very related to a wide variety of

human diseases, such as cardiovascular disease, neurodegenerative disease, inflammation,

Page 37: Computational Approach Towards Exploring the

48

allergies, diabetes, immune system dysfunctions, aging and wide variety of different cancers

(Sosa et al., 2013). Since high concentration of ROS induces damage to cell structures and

molecules including nucleic acids, proteins, lipids and membrane, from here cancer initiation and

development is linked to oxidative stress by inducing DNA mutations and DNA damage (Valko,

et al., 2006).

2.4.3 Signs and symptoms

Possible signs and symptoms include a lump, abnormal bleeding, unusual breast changes,

prolonged cough, breathlessness, unexplained weight loss, fatigue, fever, skin changes, pain, and

a change in bowel movements. While these symptoms may indicate cancer, they can also have

other causes (Tyagi, et al., 2017).

2.4.4 Cancer treatment

There are different conventional treatment modalities that are available to treat and manage

cancer. The selection of treatment and its progress depends on the type of cancer, its locality, and

stage of progression. Surgery, radiation-based surgical knives, chemotherapy, and radiotherapy

are some of the traditional and most widely used treatment methods. Some of the modern

modalities include hormone-based therapy, anti-angiogenic modalities, stem cell therapies,

targeted therapies and immunotherapy (Abbas & Rehman, 2016; Shumaila, et al., 2016).

2.4.4.1 Cancer treatment by drugs

2.4.4.1.1 Chemotherapy

Chemotherapy is considered the most effective and extensively used modality in the treatment of

most types of cancers. Out of chemotherapeutic drugs discovered, a total of 132 are FDA

approved. These drugs are designed to specifically target tumor cells and kill them by genotoxic

effect, i.e., the production of reactive oxygen species. However, chemotherapeutic drugs also

target normal cells, which could result in a variety of side effects depending on the dosage such

as hair loss, nausea, fatigue and vomiting. As a result of vigorous chemotherapy treatment,

patients become immunocompromised; this can result in complicated infections and

consequently death ( Rodgers, et al., 2012).

Different types of chemotherapy include alkylating agents, antimetabolites, anthracyclines,

mitotic inhibitors and others are mentioned in table 2.8.

Page 38: Computational Approach Towards Exploring the

49

Table ‎2.8: Various types of anticancer drugs and their examples

Types Drugs Ref.

Ch

em

oth

erap

y

Alkylating Agents

Nitrogen mustards as mechlorethamine and chlorambucil

Nitrosoureas as streptozocin and carmustine

Alkyl sulfonates as busulfan

Triazines as dacarbazine and temozolomide

Ethylenimines as altretamine

Abbas & Rehman, 2016

Antimetabolites

Pyrimidine analogue as azacitidine, capecitabine and 5-fluorouracil

Adenosine analogue as cladribine

Purine analogue as ludarabine, mercaptopurine and thioguanine

Folate analogue as mehotrexate, pemetrexed and raltitrexed

Abbas & Rehman, 2016

Anthracyclines

Daunorubicin, doxorubicin, epirubicin and idarubicin Abbas & Rehman, 2016

Mitotic inhibitors

Taxanes as paclitaxel (Taxol®) and docetaxel (Taxotere®)

Epothilones as ixabepilone (Ixempra®)

Vinca alkaloids as vinblastine, vincristine and vinorelbine

Abbas & Rehman, 2016

Topoisomerase Inhibitors as amsacrine and mitoxantrone Abbas & Rehman, 2016

Ho

rmo

na

l T

her

ap

y

Antiestrogens as tamoxifen and fulvestrant

Aromatase Inhibitors as anastrozole and letrozole

Antiandrogens as apalutamide and enzalutamide

Androgen Biosynthesis Inhibitors as abiraterone

Androgen as testosterone

Corticosteroids as dexamethasone and prednisone

Somatostatin Analogues as lanreotide

Luteinizing Hormone Releasing Hormone Agonists as buserelin

Luteinizing Hormone Releasing Hormone Antagonists as degarelix

Progestins as mroxyprogesterone and megestrol

Prolactin Lowering Agents as bromocriptine

BC Cancer Agency, 2019

Immuno-

theraputic

Agents

Cytokin as interleukin, interferon and peginterferon.

Vaccine Therapy as bacillus calmette-guerin (BCG)

Immunomodulatory drugs as lenalidomide and pomalidomide

Differentiating agents as acitretin, bexarotene and tretinoin

Ventola , et al., 2017

Targeted

Therapies

Selective kinase inhibitors

Imatinib mesylate, Gefitinib, Lapatinib, Sunitinib and Sorafenib

Monoclonal antibodies

Catumaxomab, Denosumab, Rituximab and Trastuzumab

Falzone, et al., 2018

Page 39: Computational Approach Towards Exploring the

21

2.4.4.1.2 Hormonal therapy

Rapid development in the field of molecular biology has led to the understanding of the role of

hormone in cell growth and the concept of autocrine and paracrine regulation of malignant cells.

Approximately 25% of malignant tumours in men and 40% in women have a hormonal basis.

Hormonal treatment can result in dramatic response without toxicity associated with cytotoxic

chemotherapy (EBCTC Group 2005). Examples of these drugs are given in table 2.8.

2.4.4.1.3 Immunotherapeutic agent

Cancer immunotherapy is the artificial stimulation of the immune system to treat cancer,

improving on the immune system's natural ability to fight the disease. Categories and examples

of these agents are shown in table 2.8 (Ventola, et al., 2017).

2.4.4.1.4 Targeted therapy

Targeted therapy for cancer treatment is based on tyrosine and serine/threonine protein kinase

inhibitors and monoclonal antibodies. Examples of protein kinase inhibitors include epidermal

growth factor receptor (EGFR) inhibitors, vascular endothelial growth factor receptor (VEGFR)

inhibitors, rapidly accelerated fibrosarcoma kinase (RAF) inhibitors and mammalian target of

rapamycin (mTOR) inhibitors. Monoclonal antibodies are directed toward extracellular growth

factors or extracellular receptor tyrosine kinase. Examples of these drugs are given in table 2.8

(Falzone, et al., 2018.)

2.4.4.2 Cancer treatment by natural plants

Medicinal plants are potent natural sources of drugs to treat different disease since ancient time.

The positive effect of plants in cancer treatment have been studied extensively and has shown

advanced results (Kooti, et al,. 2017). Many of anticancer lead bioactive molecules such as vinca

alkaloid, vinblastine, vincristine, camptothecin, and taxanes have been characterized from

different medicinal plants and are used as therapeutic agents worldwide (Ali, et al.,2016).

Nowadays, most research has been established to estimate the effect of medicinal plant against

cancer treatment. Numerous naturally occurring compounds from plants are being used as

anticancerous agents and are currently undergoing medical development (Malinowsky et al.,

2015). Many plants like Petasites japonicas )Malinowsky et al., 2015), Curcuma longa

(Hosseinimehr, 2014), Astragalus hamosus (Kondeva-Burdina, et al., 2014), Achillea wilhelmsii

Page 40: Computational Approach Towards Exploring the

24

(Asadi-Samani, et al., 2016), Ammi majus (Mohammed, et al., 2014), Olea europae L(Ghanbari,

et al., 2012),Thymus vulgaris L(Al-Menhali, et al., 2105), Trigonella foenum-graecum L

(Alsemari, et al., 2014) and many other plants used as an anticancerous agents with minimum

toxicity as given in table 2.9.

Table ‎2.9: Anticancerous medicinal plants

Botanical

name of plants

Family

Name

Active

components /

main parts used

Medicinal importance Ref.

Petasites

Japonicas Asteraceae Roots Anticancer Activity

Malinowsky et al.,

2015

Curcuma longa Zingiberaceae Curcumin

Activity against leukemia,

lymphoma, breast, uterus,

ovary, lung, melanoma,

colon and brain tumors

Hosseinimehr, 2014

Astragalus

Hamosus Fabaceae Aerial parts

Hepatoprotective

and antioxidant activity

Kondeva-Burdina, et

al., 2014

Silybum

marianum Asteraceae Whole plant

causes cell cycle arrest and

apoptosis

Shariatzadeh, et al.,

2014

Artemisia

vulgaris Compositae aqueous extract

Treat breast, prostate and

colon cancers. Nawab et al., 2011

Nigella sativa Ranunculaceae Seeds

Great medicinal value in

liver, colorectal, gastric and

breast cancers

Padhye et al., 2008,

Khalife, et al., 2016

Achillea

wilhelmsii Asteraceae Leaves

Cytotoxic effects on colon

cancer cells

Asadi-Samani, et al.,

2016

Allium sativum Amaryllidaceae

organosulfuric

compounds, allicin

and ajoene

reduce the risk of cancer in

breast, larynx, colon, skin,

womb, gullet, bladder, lung

and prostate cancers.

Thomson & Ali,

2003

Ammi majus Apiaceae Comorian

compounds Treat breast cancer

Mohammed, et al.,

2014

Olea europae L Oleaceae Oleuropein and

oleanolic acid

Activity against colon and

breast cancer

Ghanbari, et al.,

2012

Thymus

vulgaris Lamiaceae

Whole plant

extract

Activity against colorectal,

breast and prostate cancer

Al-Menhali, et al.,

2105

Trigonella

foenum-

graecum L

Fabaceae

Ginger, cadence,

zingerone, vanillin

and eugenol

Anticancer effects in brain

and breast cancers Alsemari, et al., 2014

In addition, Silybum marianum is used against liver cancer and shown tumorigenic effects in-

vitro and in-vivo conditions by suppressing oxidative stress and proliferation (Shariatzadeh, et

al., 2014). Nigella sativa has been shown several pharmacological activities, including

antioxidant, anti-inflammatory, chemotherapeutic and antitumor activities (Padhye, et al., 2008),

as well as hepatoprotective activity (Khalife, et al., 2016). Artemisia vulgaris aqueous extract has

anticancer activity against human breast carcinoma, human prostate cancer and colon cancer

(Nawab, et al., 2011). Various researches have shown that Allium sativum, organosulfuric

Page 41: Computational Approach Towards Exploring the

22

compounds and allicin reduce the risk of cancer in breast, larynx, colon, skin, womb, gullet,

bladder, prostate and lung (Thomson, et al., 2003).

2.5 Diabetes mellitus (D.M)

Diabetes is a serious, chronic disease that occurs either when the pancreas does not produce

enough insulin (a hormone that regulates blood glucose), or when the body cannot effectively

use the insulin it produces. Raised blood glucose, a common effect of uncontrolled diabetes by

over time, lead to serious damage to the heart, blood vessels, eyes, kidneys and nerves (WHO,

2017).

Diabetes mellitus is the seventh leading cause of death globally. In 2019, it is estimated that,

approximately (one in 11) 463 million adults (20-79 years) were living with diabetes; by 2045

this will rise to 700 million. About 79% of adults with diabetes were living in low- and middle-

income countries. One in 2 (232 million) people with diabetes were undiagnosed. More than 1.1

million children and adolescents are living with type 1 diabetes. Ten Percentage of global health

expenditure is spent on diabetes (International Diabetes Federation (IDF), 2019).

The Palestinian Ministry of Health (PMoH) ranked T2DM as the fourth leading cause of death

and represented 8.9% of all deaths in 2014 (PMoH, 2016).

The International Diabetes Federation (IDF) reported the prevalence in diabetic patients aged

20–79 years in Palestine to be 9.1% .The percentage of DM between both sexes is equal. The

percentage of patients with T1DM is 4.7% and 95.3% of T2DM (IDF, 2015).

2.5.1 Classification of diabetes mellitus

Type 1 diabetes mellitus (T1DM) is an autoimmune disease that results in β cell destruction. It

comprises about 5%–10% of total cases of diabetes. It is typically recognized in childhood or

adolescence. It is used to be known as juvenile-onset diabetes or insulin dependent diabetes

mellitus (IDDM). It is associated with the presence of islet cell antibodies, and patients require

lifelong insulin (Ashcroft & Rorsman, 2012).

T2DM is a non–insulin dependent (NIDDM). It is due to insulin resistance or reduced insulin

sensitivity, combined with relatively reduced insulin secretion which in some cases becomes

absolute. The body tries to overcome this resistance by secreting more and more insulin. At least

90% of patients with diabetes have T2DM (Mau, et al., 2019).

Page 42: Computational Approach Towards Exploring the

22

Gestational diabetes is a form of diabetes that start during the second half of pregnancy and

produced by the hormones of pregnancy or by a shortage of insulin. Women who have

gestational diabetes are more likely than other women to have large babies and to develop type II

diabetes later in life. However, such kind of diabetes resolves as the nannies delivers (Mau, et al.,

2019).

2.5.2 Signs and symptoms

The classic symptoms of untreated diabetes are polyuria (increased urination), polydipsia

(increased thirst), polyphagia (increased hunger) and unintended weight loss. Symptoms may

develop rapidly (weeks or months) in type 1 diabetes, while they usually develop much more

slowly and may be subtle or absent in type 2 diabetes. Other Signs and Symptoms include

blurred vision, headache, fatigue, irritability, slow healing of cuts and itchy skin (WHO, 2019).

2.5.3 Complications of diabetes mellitus

Raised blood glucose can result in multiple complications as diabetic retinopathy that may

develop into loss of vision, end stage renal disease, cardiovascular events and lower extremities

amputation (WHO, 2017).

2.5.4 T2DM pathogenesis and major risk factors

The pathogenesis of T2DM is sophisticated and its corresponding mechanisms remain

conflicting. T2DM was generally considered as chromosome polygene recessive inheritance,

following with abnormality of insulin secretion (Morris, et al., 2012). Molecularly, obvious

alterations occurred in insulin genes compared with normal patients, suggesting that changes of

insulin genes may be one of contributing factors in the pathogenesis of T2DM (Scott, et al.,

2012).

With regard to the environment, various kinds of bacteriostatic agents, preservatives, antibiotics,

chemicals abuses and even lifestyle along with irregular dietary habits are external factors which

are always neglected (He, et al., 2019).

In T2DM, insulin resistance contributes to increased glucose production in the liver and

decreased glucose uptake in muscle and adipose tissue. In addition, β-cell dysfunction results in

reduced insulin release, which is insufficient for maintaining normal glucose levels. Indeed,

Page 43: Computational Approach Towards Exploring the

21

multiple defects contribute to the development of glucose intolerance and hyperglycemia in

T2DM as shown in figure 2.3 (DeFronzo, et al., 2014).

Figure ‎2.3: Multiple defects contribute to the development of glucose intolerance in T2DM. HGP, Hepatic

glucose production (DeFronzo, et al., 2014).

Importantly, dysfunction of glucose metabolism, incretin secretion and insulin sensitivity

mentioned with some key enzymes such as α-glucosidase, α-amylase, dipeptidyl peptidase 4

(DPP-4), peroxisome proliferator-activated receptor-γ (PPAR-γ), protein tyrosine phosphatase1

B (PTP1B) and glucose transporter-4 (GLUT-4) quietly account for the pathogenesis of T2DM

(He, et al., 2019).

Pharmacologically, α-amylase hydrolyzes macromolecules like starch into some oligoglucans.

These substances are further degraded by α-glucosidase into absorbable glucose at the brush

border of small intestine and then permeate blood circulation. Elevated blood glucose increase

glucagon-like peptide-1 (GLP-1) production to enhance insulin release and protect pancreatic β-

cells. DPP-4 can shorten the half-life of GLP-1,which interrupts glucose metabolism (Liu, et al.,

2017).

Page 44: Computational Approach Towards Exploring the

25

Insulin secreted by pancreatic β-cells can modulate lipid metabolism and hepatic and muscle

glycogen synthesis, which require the regulation of GLUT-4, PTP1B and PPAR-γ pathways (He,

et al., 2019).

2.5.5 Management of T2DM

2.5.5.1 Non pharmacological treatment

Modification of lifestyle, including weight loss, increasing physical activity and adopting a

healthy diet, remains one of the first line strategies for the management of T2DM (Zheng, et al.,

2018).

2.5.5.2 Pharmacological treatment

Current remedies for T2DM mainly include chemical or biochemical agents such as biguanides,

sulfonylureas, α-glucosidase inhibitors, etc. All these first line clinical therapeutic drugs and

corresponding mechanisms of action are shown in table 2.10 (He, et al., 2019).

Table ‎2.10: Marketed therapeutic agents for T2DM and respective mechanisms (He, et al., 2019)

Categories Familiar drugs Mechanism Adverse Effects

2nd

Generation

Sulfonylureas

Glibenclamide Gliclazide,

Glimepiride and Glipizide

Stimulation of pancreatic

insulin secretion

Hypoglycemia risk, Weight

gain

α-Glucosidase

INHIBITORS

Acarbose, Miglitol

Vocarbose

Disturbance of glucose

digestion and absorption in

the intestinal system

Gastrointestinal symptoms

Biguanides Phenformin and

Metformin

Increases insulin sensitivity

and hepatic glucose

utilization

Gastrointestinal symptoms,

Lactic acidosis, contraindi-

cated in renal insufficiency

DPP-4 Inhibitors Linagliptin, Sitagliptin

Saxagliptin, Vildagliptin

And Alogliptin

Decrease glucagon and

blood glucose levels

through elongating the half-

life of GLP-1

Nasopharyngitis, Headache,

Nausea and Hypersensitivity

Thiazolidinediones Pioglitazone and

Rosiglitazone

Amelioration of insulin

action by stimulating of the

PPAR-γ

Hepatoxicity, Weight gain,

Risk for heart failure, Fluid

retention

GLP-1 Agonists Exenatide , Liraglutide

Exenatide, and Albiglutide

Promotion of insulin

signaling; inhibition of

elevated glucagon levels

Risk for thyroid cancer

SGLT-2 Inhibitors

Canagliflozin,

Dapagliflozin and

Empagliflozin

Decrease of glucose

secretion in the renal organ

Genital infections and

Weight loss

DPP-4: Dipeptidyl peptidase-4, GLP-1: Glucagon-like peptide-1, SGLT2: Sodium-glucose cotransporter-2

Page 45: Computational Approach Towards Exploring the

26

2.5.5.3 Diabetes management by natural plants

Fortunately, natural occurring antidiabetic drugs have never been obsoleted and still play a

principal role in the management of T2DM, many of which are known to be effective against

diabetes (Bailey & Day, 2004).

Academically, He et al., reviewed the hypoglycemic effects of many plants, their identified

compounds, crude extracts and their molecular targets for treating T2DM from 2011 to 2017

(He, et al., 2019). Some of these plants are listed in table 2.11.

Table ‎2.11: Medicinal plants used in the treatment of T2DM (He, et al., 2019)

Target Plant specie Familly Part used Active ingredient

α-

Glucosidase

and α-

Amylase

Allium sativum L Alliaceae Ground bulbs Kaempferol , Luteolin and Quercetin 4'-glucoside

Salvia offcinalis L Lamiacea Leaf Luteolin and Quercetin4'-glucoside

Momordica charantia Cucurbitacea Fruit Crude extract

Swertia mussotii Gentianacea Whole plants Xanthones

Zingiber mioga Gentianaceae Whole plants Ethyl alcohol and water extracts

DPP-4

Abelmoschus

esculentus Malvacea Root

Quercetin, Glucosides and

triterpenes

Apocynum venetum Apocynaceae Leaf Isoquercitrin

Origanum vulgare Lamiaceae Seed Naringenin hispidulin, Cirsimaritin

and Carnosol

Pericarpium Citri

Reticulatae Rutaceae Peels Naringin

Pilea microphylla Urticaceae Wholeplants Flavonoids

PPAR-γ

Ampelopsis

grossedentata Vitaceae

Stem

Leaf Ampelopsin

Citrus junos Rutaceae Peel Pulp Ethanol extract of the pulp

Glycyrrhiza inflata Leguminosae Root Licochalcone E

Morinda citrifolia Rubiaceae Root Ethanolic extract

Opuntia humifusa Cactaceae Stem Flavonoids

PTP1B

Annona squamosal Ranunculaceae Fruit Hexane extract

Dodonaea viscosa Sapindaceae Aerialparts Polyphenolic compounds

Persea americana Lauraceae Lea Aqueous extract

Ramalina Americana Ramalinaceae Bark Trivaric acid

Syzygium cumini Myrtaceae Seeds Vitalboside

GLUT-4

Catharanthus roseus Apocynaceae Leaf Ethanolic extrac

Centratherum

anthelminticum Asteraceae Seeds Methanolic extract

Psoralea corylifolia Fabaceae Seeds Bavachin

Rosmarinus offcinalis Lamiaceae Leaf Rosmarinic acid

Zingiber officinale Zingiberaceae Root Ethyl acetate extract

DPP-4: Dipeptidyl peptidase-4, GLUT4: Glucose transporter 4, PPAR-γ: Peroxisome proliferator activated receptor-γ, PTP1B: Protein tyrosine phosphatase 1B.

Page 46: Computational Approach Towards Exploring the

27

2.6 Hyperlipidemia

Hyperlipidemia is an increase in one or more of the plasma lipids, including triglycerides,

cholesterol, cholesterol esters and phospholipids and or plasma lipoproteins including very low

density lipoprotein and low-density lipoprotein as well as a reduce in high-density lipoprotein

levels (Shattat, 2014).

2.6.1 Classification of hyperlipidemia

Hyperlipidemia can be claasified into primary hyperlipidemia (also called familial) and

secondary Hyperlipidemia. Primary hyperlipidemia takes place as a result of genetic problems

i.e., mutation within receptor protein, which may be due to single (monogonic) gene defect or

multiple (polygenic) gene defect. Secondary hyperlipidemia arises as a result of other

underlining diseases like diabetes, myxoedema, nephritic syndrome, chronic alcoholism and with

use of drugs like corticosteroids, oral contraceptives and beta blockers (Pe, et al., 2015).

2.6.2 Causes and risk factors of hyperlipidemia

The causes of hyperlipidemia include genetic factors with risk factors like excessive alcohol

consumption, obesity, medications (hormones or steroids), diabetes, metabolic syndrome, long

term kidney disease, premature menopause, an underactive thyroid gland, or hypothyroidism,

pregnancy and sedentary lifestyle (Singh & Nain, 2018).

2.6.3 Complications of hyperlipidemia

Hyperlipidemia increases morbidity and mortality when combined with other prevalent diseases

such as diabetes mellitus, hypertension and cardiovascular diseases and it may leads to several

harmful diseases like atherosclerosis, cardiovascular diseases, high blood pressure and many

other severe problems which seriously affect the human body (Singh & Nain, 2018).

2.6.4 Pathophysiology of hyperlipidemia

The pathophysiology of primary hyperlipidemia involve the idiopathic hyperchylomicronemia in

which defect in lipid metabolism leads to hypertriglyceridemia and hyperchylomicronemia

caused by a defect in lipoprotein lipase (LPL) activity or the absence of the surface apoprotein

CII31 (Gotto & Moon, 2010; Amit, et al., 2011).

Page 47: Computational Approach Towards Exploring the

28

In secondary hyperlipidemia, the postprandial absorption of chylomicrons from the

gastrointestinal tract occurs 30- 60 min after ingestion of a meal containing fat that may increase

serum triglycerides for 3-10 hours. The diabetes mellitus patients have been noted to possess low

LPL activity which further caused high synthesis of very low density lipoprotein (VLDL)

cholesterol by the liver ultimately leading to hyperlipidemia. Moreover, hypothyroidism induced

low LPL activity and lipolytic activity has been noted to reduce hepatic degradation of

cholesterol to bile acids. Furthermore, hyperadrenocorticism increased the synthesis of VLDL by

the liver causing both hypercholesterolemia and hypertriglyceridemia (Pe, et al., 2015).

2.6.5 Management of Hyperlipidemia

2.6.5.1 Non pharmacological management

Stress reduction, dietary modification, reduce the risk factors of atherosclerosis like body weight,

consumption of alcohol, smoking and treatment of diseases like hypothyroidism, DM,

hypertension are also important while starting hypolipidemic drug therapy (Atlee, 2020).

The objectives of dietary therapy are to decrease the intake of total fat, saturated fatty and

cholesterol progressively and to achieve a desirable body weight. The dietary therapy includes

reduction of saturated fat intake to 7 percent of daily calories, reduction of total fat intake to 25

to 35 percent of daily calories, reduction of dietary cholesterol to less than 200 mg per day,

eating 20 to 30 g a day of soluble fiber, which is found in peas, beans, and certain fruits and as

well as increasing the intake of plant stanols or sterols, substances found in nuts, vegetable oils,

corn and rice, to 2 to 3 g daily. Other foods that can help control cholesterol include cold-water

fish, such as mackerel, sardines, and salmon. These fish contain omega-3 fatty acids that may

lower triglycerides. Soybeans found in tofu and soy nuts and many meat substitutes contain a

powerful antioxidant that can lower LDL (Arun, et al., 2013).

2.6.5.2 Pharmacological therapy

Generally, the drugs involves in the treatment of hyperlipidemia are classified into several

groups such as statins, resins, fibric acid derivatives, niacin and novel drugs as shown in table

2.12 (Arun, et al., 2013; Atlee, 2020)

Page 48: Computational Approach Towards Exploring the

29

Table ‎2.12: Antihyperlipidemic drugs and their mechanisms of action (Atlee, 2020)

Drug Mechanism 0f action Side effects Effects on lipid

HMG-CoA reductase

inhibitors (Statins):

Lovastatin(20-40mg/day)

Simvastatin(5-20mg/day)

Atorvastin(10-40mg/day)

Rosuvastin(5-20mg/day)

↓ cholesterol biosynthesis

by inhibiting HMG-CoA

Rise serum transaminase

and creatine levels,

Muscle Tenderness,

Myopathy(rare)

↓LDL,

↑HDL and

↓TG

Ezetimibe (5-10 mg/day) ↓ Cholesterol absorption Contraindicated in

pregnancy and children ↓LDL

Bile acid sequestrants (Resins)

Cholestyramine(4-16mg)

Colestipol (5-30mg)

↓ bile acid absorption and

↑hepatic conversion

of cholesterol to bile acids

Nupalatable,

Flatulence,

Interference with drugs

↓ LDL and

↑HDL

Fibric acid derivatives

Gemfibrozil(1200mg)

Benzafibrate (600mg)

Fenofibrate(200mg)

↓ PPAR-α, ↑ activity of

LPL, ↓ release of FA from

adipose tissue

Skin rashes, Eosinophillia

Impotency, contraindica-

ted in pregnancy.

↓LDL,

↑HDL and

↓TG

Tesaglitazar ↓ PPAR-α

↓ PPAR-γ

Gastrointestinal

symptoms,

Respiratory infections

↓LDL, ↑HDL

And ↓TG

Niacin (2-8 gms/day) ↓ lipolysis in adipocytes,

↓ FA synthesis in liver

↓ VLDL

Flushing,

Itching

↓LDL, ↓TG

And ↑HDL

Novel drugs

PCSK9 inhibitors

Alirocumab

Evolocumab

PCSK9 promote LDL

receptor destruction on liver cells, prevent blood

clearance of LDL

.

Chest pain,fluid retention,

Hepatic steatosis, itching and irritation

↓ LDL

MTP inhibitors

Lomitapide

↓ Assembly of

apolipoproteins,

triglycerides and

cholesterol in liver

Hepatic steatosis and

Increase in liver

transaminase level are the

serious side effects

↓ LDL and

↓ TG

Bempedoic Acid ↓ACL (links carbohydrate

metabolism to pathways

for synthesis of Fas and

cholesterol).

Muscle pain, diarrhea and

pain in extremities

↓ Cholesterol

↓ FA and ↓LDL

ACAT: Acyl coenzyme A cholesterol acyl transferase, ACL: adenosine triphosphate citrate lyase, FA: fatty acid,

HMG-CoA: Hydroxy-3-Methylglutaryl Coenzyme A, HLD; High density lipoprotein, LDL ;Low density

lipoprotein, LPL: lipoprotein lipase, MTP: Microsomal Triglyceride transfer Protein, PCSK9: Proprotein

Convertase subtilisin/ kexin type 9, TG: Triglycerides, VLDL: Very Low density lipoprotein.

2.6.5.3 Management of hyperlipidemia by natural plants

Over the past decade, natural plants have become a topic of global importance, making an impact

on the world health. Natural plants continue to play a central role in the healthcare system of

large proportions of the world‘s population. Currently used hypolipidemic drugs are associated

with so many adverse effects which are not seen with herbal preparations. Plant parts or plant

Page 49: Computational Approach Towards Exploring the

21

extract are sometimes even more potent than known hypolipidemic drugs. Some plants which

had hypolipidemic activity are presented in table 2.13 (Arun, et al., 2013).

Table ‎2.13: Plants with hypolipidemic activity

Plant Family Parts used Ref

Amaranthus spinosus Amaranthaceae Leaves Arun, et al., 2013.

Glycyrrhiza glabra Fabaceae Root Arun, et al., 2013.

Withania somnifera Solanaceae Root Arun, et al., 2013.

Chlorophytum

borivilianum Liliaceae Root Arun, et al., 2013.

Moringa oleifera Moringaceae Leaves ,root, seeds Arun, et al., 2013.

Hibiscus cannabinus Malvaceae Fresh leaves Arun, et al., 2013.

Randia dumetorum Rubiaceae Fruit Arun, et al., 2013.

Medicago sativa Fabaceae Seeds Bahmani, et al., 2015.

Trigonella foenum-

graecun Fabaceae Seeds Bahmani, et al., 2015.

Allium sativum L Amaryllidaceae Alliin Bahmani, et al., 2015.

Silybum marianum L Asteraceae Silymaryne Bahmani, et al., 2015.

2.7 Computer aided drug design(CADD(

Drug discovery is challenging, an exhaustive and time-consuming process involving numerous

stages like target identification, validation, lead optimization, preclinical trials, clinical trials and

finally post marketing vigilance for drug safety. For instance, the total average cost of

developing a new drug, as per an estimate, ranges from $2 billion to $3 billion and it takes at

least 13–15 years to bring a drug to the market—starting from initial discovery to the approval

stage. The application of computer aided drug designing (CADD) is an indispensable approach

for developing safe and effective drugs. It is extensively used to reduce cost, time and speed up

the early stage development of biologically new active molecules (Scannell, et al., 2012).

CADD based approaches including pharmacophore modeling, molecular docking, inverse

docking, chemical similarity (CS), quantitative structure activity relationship (QSAR), virtual

screening (VS) and molecular dynamics simulations have been quite productive in predicting the

therapeutic outcome of candidate drugs/compounds besides saving precious time. Computational

approaches have led to the discovery of many drugs that have passed preclinical and clinical

trials and become novel therapeutics in the treatment of a variety of diseases. In addition, CADD

plays an important role in predicting absorption, distribution, metabolism, excretion and toxicity

(ADME/T) of candidate drugs. Overall, CADD represents an effective and much-needed strategy

for designing therapeutically effective drugs to combat human diseases (Dar, et al., 2018).

Page 50: Computational Approach Towards Exploring the

24

In a drug discovery campaign, CADD is usually used for three major purposes; filter large

compound libraries into smaller sets of predicted active compounds that can be tested

experimentally, guide the optimization of lead compounds, whether to increase its affinity or

optimize drug metabolism and pharmacokinetic (DMPK) properties including absorption,

distribution, metabolism, excretion and the potential for toxicity (ADME/T) and design novel

compounds, either by "growing" starting molecules one functional group at a time or by piecing

together fragments into novel chemotypes. Figure 2.4 illustrates the position of CADD in drug

discovery pipeline (Sliwoski, et al., 2013).

2.7.1 CADD classification

CADD methods can be broadly classified into two groups, namely structure based (SB) and

ligand based (LB) drug discovery as shown in figure 2.4 The CADD method used depends on

the availability of target structure information (Leelananda & Lindert, 2016; Pares, et al., 2017).

In order to use SBDD tools, information about target structures needs to be known. Target

information is usually obtained experimentally by X-ray crystallography or NMR (nuclear

magnetic resonance). When neither is available, computational methods such as homology

modeling may be used to predict the three dimensional structures of targets. Knowing the

structure makes it possible to use structure-based tools such as virtual high throughput screening

and direct docking on targets and possible drug molecules (Leelananda & Lindert, 2016).

The central goal of SBDD is to design compounds that bind tightly to the target, e.g., with large

reduction in free energy, improved DMPK/ADME/T properties, and are target specific

(Jorgensen, 2010).

When the target structure is not experimentally determined or it is not possible to predict the

structure using computational methods, ligand-based approaches are often used as an alternative

methods. These methods, however, rely on the information about known active binders of the

target. Pharmacophore modeling, molecular similarity approaches and QSAR modeling are some

popular LBDD approaches (Leelananda & Lindert, 2016).

Page 51: Computational Approach Towards Exploring the

22

Figure ‎2.4: CADD in drug discovery/design pipeline (Leelananda & Lindert, 2016).

2.7.2 Computational target fishing

Computational or in-silico target fishing which is also known as reverse screening as seen in

figure 2.5 has emerged as an interdisciplinary field with tremendous potential to advance in-

silico drug design. Target fishing, or target identification, is an important start step in modern

drug development, which investigates the mechanism of action of bioactive small molecules by

identifying their interacting proteins. It can also be used to find potential off-targets of

therapeutic compounds for the study of their side effects. In addition, target fishing can be used

to detect drug polypharmacology (Achenbach, et al., 2011) and for drug repurposing, (Liu, et al.,

2013).

Computer Aided Drug Design (CADD)

Target Identification

Structure Based Drug

Design (SBDD):

Structure prediction

Docking

De novo ligand design

Ligand Based Drug

Design (LBDD):

QSAR

Pharmacophore modeling

Similarity search

In Vitro Verification

Drug Candidate

Hit

Lead generation

Page 52: Computational Approach Towards Exploring the

22

Figure ‎2.5: Comparison between docking and reverse docking (Zheng et al., 2013).

2.7.2.1 Polypharmacology

Polypharmacological phenomena includes single drug acting on multiple targets of a unique

disease pathway, or single drug acting on multiple targets pertaining to multiple disease

pathways. In addition, polypharmacology for complex diseases is likely to employ multiple

drugs acting on distinct targets, that are part of a networks regulating various physiological

responses (Reddy & Zhang, 2013). The polypharmacological approaches aim to discover the

unknown off-targets for the existing drugs (also known as drug repurposing) (Achenbach, et al.,

2011). The approach needs the systematic integration of the data derived from different

disciplines including computational modeling, synthetic chemistry, in vitro / in vivo pharmacolo-

gical testing and clinical studies (Reddy & Zhang, 2013).

Almasri, 2018, used ROCS based target fishing approach (RTFA) to approve the

polypharmacology of some natural products as resveratrol, curcumin and berberine, he explored

some targets that were reported previously and other targets which weren't reported. In his study

docking was adopted for the unreported targets and the binding energy was calculated, e.g.,

resveratrol, among other compounds involved in the study, is non-flavonoid polyphenol has

antioxidant, phytoestrogenic, anti-inflammatory, anticarcinogenic, antiaging, cadioprotective and

neuroprotective activities. The exact mechanisms of action of some of these potential biological

activities wern't fully clarified, at his work, he discovered the targets that clarify these activities

Page 53: Computational Approach Towards Exploring the

21

as estogen receptor α (ERα) for anti-inflammatory activity, silent information regulator proteins-

1 (SIRT-1) which responsible for antiobesity, antiaging and anticancer activities, protein kinases

as casein kinase 2 (CK2) and (PIM1) for anticancer activity, in addition, resveratrol was found to

act on tankyrase 2 as anticancer target. Moreover, resveratrol captured two targets of

neuroprotective properties that were transthyretin and β-secretase (Almasri, 2018).

2.7.2.2 Drug repurposing

Drug repurposing (also known as repositioning, reprofiling, or rediscovering) is defined as

developing new uses for a drug beyond its original use or initial approved indication (Ashburn &

Thor, 2004; Mangione, et al., 2019).

The premise is that since most approved compounds have known bioavailability and safety

profiles, proven formulation and manufacturing routes, and reasonably characterized

pharmacology, repositioned drugs can enter clinical phases more rapidly and at a lower cost than

novel compounds. It is therefore not surprising that in recent years, of the new drugs that reach

their first markets, repositioned drugs have taken up to a percentage of approximately 30%. For

instance, of the 113 new drugs approved or launched in 2017, only seven were first-in-class

agents (an approved and launched first drug with a novel mechanism of action) while 36 were

repositioned drugs (Papapetropoulos & Szabo, 2018).

Most of the successful cases of drug repurposing have been serendipitous discoveries rather than

systematic, hypothesis-driven outcomes. There are many stories of repurposing that have gone

on to be profitable: bupropion, originally used for depression, was repurposed for smoking

cessation; and thalidomide, used as a treatment for morning sickness, is now used for multiple

myeloma (Baker, et al., 2018).

In addition, scientists from the University of Dundee have shown that the antitubercular drug

delamanid has the potential to be repositioned as an oral drug for visceral leishmaniases, one of

the major diseases seen in developing countries (Patterson, 2016).

In addition, Kinnings, et al., 2009, performed extensive structure-based studies on nine different

Mycobacterium tuberculosis InhA structures (InhA: 2-trans-enoyl-acyl carrier protein reductase

which is a target of the antituberculosis isoniazid) to evaluate whether the entacapone and

tolcapone drugs, approved for the treatment of parkinson‘s disease, might be repurposed against

tuberculosis. Their results allowed the identification of entacapone as a promising lead

compound against resistant strains of Mycobacterium tuberculosis (Kinnings, et al., 2009).

Page 54: Computational Approach Towards Exploring the

25

On the same line, Dakshanamurthy, et al., 2012, performed extensive docking-based virtual

screenings on a subset of compounds taken from Protein Drug Bank (PDB) and FDA databases

against several X-ray crystal structures of human proteins reported in the PDB. According to the

reported results, the authors discovered that the antiparasitic drug mebendazole is also an anti-

angiogenic VEGFR2 inhibitor. Moreover, they also successfully discovered that the COX-2

inhibitor celecoxib and dimethyl celecoxib bind to Cadherin-11, which is a protein mediating

calcium-dependent cell-cell adhesion that plays a crucial role in rheumatoid arthritis

(Dakshanamurthy, et al., 2012).

2.7.2.3 Computational methods for target fishing

Various computational methods have been developed to predict the molecular targets of a

compound. These methods were initially classified into four groups: molecular similarity

searching, data mining/machine learning, panel docking, and the analysis of bioactivity spectra.

Also, other classes, such as protein-structure-based methods, have been proposed (Zheng, et al.,

2013; Cereto-Massagué, et al., 2015).

2.7.2.3.1 Molecular similarity methods

This section describes chemical similarity methods and shape based similarity methods. The

simplest methods for target prediction are based on chemical similarity and the use of current

knowledge about the bioactivity of millions of small molecules. These methods are based on the

‗‗chemical similarity principle,‘‘ which states that similar molecules are likely to have similar

properties. Thus, the targets of a molecule can be predicted by identifying proteins with known

ligands that are highly similar to the query molecule. The advantage of these methods is that they

only require the computation of the similarity between compounds. In the chemical similarity

method, a small molecule is represented as a chemical fingerprint. Fingerprints are a way of

encoding the structure of a molecule. The most common type of fingerprint is a series of binary

digits (bits) that represent the presence or absence of particular substructures in the molecule. To

compare the fingerprints of two molecules, the Tanimoto coefficient or any other similarity

criterion can be used. The more similar two compounds are, the closer the Tanimoto coefficient

will be to 1. (Cereto-Massagué, et al., 2015).

Tanimoto coefficient also known as the Jaccard coefficient, one of the most common approaches

for database searching due to its simplicity, fast speed as its calculation does not involve any

square root, making it faster in calculation, easy implementation and results in drug discovery

(Yu, et al., 2015). Tanimoto binary variable formula for binary data, as shown in the equation:

Page 55: Computational Approach Towards Exploring the

26

When the molecules A and B, a = bits set to 1 in A, b = bits set to 1 in B, c = number of 1 bits

common to both (Himmat, et al., 2016).

Molecular similarity can be classified into two Dimension (2D) chemical similarity and three

Dimension (3D) molecular similarity.

The 2D fingerprints have been widely used for similarity searching in target fishing due to quick

calculation so in some cases, methods that use 2D fingerprints outperform those methods that use

3D fingerprints in correct target prediction (Nettles, et al., 2006). The 3D chemical descriptors

can also be used for similarity searching in target fishing, although calculating them is

computationally more expensive. Because they contain more information, the predictions based

on 3D fingerprints would be expected to be better than those based on 2D fingerprints. The 3D

descriptors work better in cases of low structural similarity (Nettles, et al., 2006).

3D similarity searching requires the conformation properties and the specification of an entire

target structure (the whole structure) rather than partial structure. Thus, the 3D method-based

similarity measurement was introduced and gained more attention recently because of their

potential to overcome the key limitation of 1D and 2D methods (Nettles, et al., 2006).

The 3D structures usually have three different angles of view, which is x-, y-, and z- axis. With

the one extra angle of view in 3D structure, it will provide an important information needed and

better conformation for the similarity search process. Another characteristic of 3D structure is, it

is better in capturing all aspects of the molecular structure (the size and the shape of molecular

structure). In order to measure how similar is the chemical structure is, a similarity measure is

needed (Cereto-Massagué et al., 2015).

Shape-based similarity methods use 3D shape comparisons between molecules, usually

comparing the shape of the molecular volume, but other ―shapes‖ can be compared, like the

electrochemical surface. This can be done with software such as ROCS, Phase Shape,

ESHAPE3D, PARAFIT, ShaEP and USR as some examples (Cereto-Massagué, et al., 2015)

Popular 3D similarity comparison programs (Shape-it, Align-it, and ROCS). These programs

aligned molecules based on either molecular shape (Shape-it and ROCS) or pharmacophore

(Align-it) features using a Gaussian-like function. CSNAP (chemical similarity network analysis

pull-down) for drug target profiling using chemical similarity networks.

Tanimoto binary variable formula: SA,B = c \ a+b-c

Page 56: Computational Approach Towards Exploring the

27

A known limitation of chemical similarity approaches is that this approach often suffers from

false positive and false negative predictions, particularly when inactive and active compounds

display structural similarities. In many cases (where there is no known biologically active

conformation for the molecule), a single low-energy conformer is used, although it can be

biologically irrelevant. Another approach is to get the conformation of the molecules by aligning

them to a known bioactive conformation of a known ligand. In other cases, combining chemical

and shape similarity measures significantly increases the target prediction accuracy (Gfeller, et

al.,2014).

In addition, statistical analysis has been added to traditional chemical similarity scores in order to

assess the statistical significance of similarity. For example, fitting with extreme value

distributions, the similarity ensemble approach models the possibility of the occurrence of higher

scores when comparing two ligand sets. This method has been successfully used in drug

repurpose and side effect prediction (Wang, et al., 2013).

Rapid overlay of chemical structures software (ROCS)

ROCS is a standard tool for the calculation of 3D shape and chemical (‗‗color‘‘) similarity.It is a

powerful virtual screening tool which can rapidly identify potentially active compounds by

shape/chemical matching between the natural compounds and the chemogenomic database.

ROCS is competitive with, and often superior to, structure-based approaches in virtual screening,

both in terms of overall performance and consistency. Novel and interesting molecular scaffolds

have been identified using ROCS against targets often considered very difficult for

computational techniques to address. ROCS software is designed to perform large scale 3D

database searches by using a superposition method that finds the similar but non-intuitive

compounds that are so valuable in the drug discovery process (Hawkins et al., 2007; ROCS

3.2.1.4, 2015; Kearnes and Pande, 2016).

ROCS is a shape-based superposition method, for definition of shape. Two entities will have the

same shape if their volumes exactly correspond. The more the volumes differ, the more the

shapes will differ but a volume is any scalar field. The special case for the common

understanding of volume is a specific scalar field that has a value of one inside an object and

zero outside (ROCS 3.2.1.4, 2015).

Page 57: Computational Approach Towards Exploring the

28

ROCS uses only the heavy atoms of a ligand, hydrogens are ignored. Since shape and volume in

this context are so closely related, a volume overlap maximization procedure is an excellent

method for gaining insights into similar shapes (ROCS 3.2.1.4, 2015).

Although ROCS is primarily a shape-based method, user specified definitions of chemistry

alignment, known as ‗color‘ can be included into the superposition and similarity analysis

process which facilitates the identification of those compounds which are similar both in shape

and chemistry (Hawkins, et al., 2007).

ROCS can routinely perform global shape and color alignments at the rate of 600-800

conformers per second (Kearnes and Pande, 2016).

Molecules are traditionally viewed as a set of fused spheres. Molecules are aligned by a solid

body optimization process that maximizes the overlap volume between them as seen in figure

2.6. Volume overlap in this context is not the hard-sphere overlap volume, but rather a Gaussian-

based overlap parameterized to reproduce hard-sphere. (Kearnes and Pande, 2016).

Figure ‎2.6: Molecular descriptors based on the ROCS color force field. Color components represent each

colortype independently. Color atom overlaps describe similarity in terms of individual color atoms in the query

molecule (bottom left). Color component values are Tanimoto scores. Note that color atom overlap volumes are

used without normalization, and that negative values indicate favorable interactions (Kearnes and Pande, 2016).

Medium-sized database searching (Ten‘s of millions of conformers) becomes tractable but slow

at this rate of superposition. Distributed computing makes the entire process much more facile

Page 58: Computational Approach Towards Exploring the

29

for screening larger numbers of compounds and conformers. ROCS can automatically split up

similarity searches over entire networks of computers in an efficient and manner taking full

advantage of parallel virtual machines. The coupling of shape and chemistry screening with a

distributed architecture makes ROCS an incredibly powerful tool for searching large 3D

databases (ROCS 3.2.1.4, 2015).

ROCS includes a query visualizer and editor, vROCS, which provides a graphical interface for

creating queries and evaluating the performance of ROCS (Kearnes and Pande, 2016).

Items used in ROCS Report

Name: this is the name of the database molecule. If the database contains multi-conformer

molecules, the specific conformer index is appended to the molecule name with an underscore.

ShapeQuery: This is the name of the query molecule. If the query is a multi-conformer

molecule, then the specific conformer index is appended to the molecule name with an

underscore.

Rank: It is the numerical ranking in the hit list, based on the chosen score to sort by.

TanimotoCombo: To provide a score that includes both shape fit and color, the Shape Tanimoto

is added to the Color Tanimoto, resulting in the TanimotoCombo score. This has a value between

0 and 2 and is the score used for ranking the hit list.

Shape Tanimoto: This column gives the ShapeTanimoto, a value between 0 and 1 as calculated

by the Tanimoto equation.

Color Tanimoto: This column gives the ColorTanimoto, a value between 0 and 1 as calculated

by the Tanimoto equation (Kearnes and Pande, 2016).

2.7.2.3.2 Data mining and machine learning methods

In data-mining or machine learning methods, the properties of known active compounds against

a target are analyzed carefully and statistical models are generated, which after rigorous training

are employed to predict the probable targets that associate with the query compound. The main

limitation of this method is that every target may bind structurally diverse classes of compounds

and hence one model may not cover all the features, consequently affecting the performance in

target fishing (Wang, et al, 2013; Ganesan & Barakat, 2016).

Page 59: Computational Approach Towards Exploring the

11

2.7.2.3.3 Molecular docking methods

Other computational target fishing methods use the protein structure of the targets to predict

novel bioactivities. Pharmacophore searching, protein−ligand interaction fingerprints or protein

docking can be used. These methods are limited to targets with resolved structures. Molecular

docking methods for target fishing employs a ‗reverse‘ virtual screening approach, in which a

compound of interest is docked into a wide array of protein structures in public databases as seen

in figure 2.5, such as protein data bank (PDB), and the target in the best scoring complex is

predicted to be a probable partner of the query compound. Several online servers, such as

TarFisDock, INVDOCK and idTarget, have been developed for this purpose. However, the

accuracies of these docking-based methods are dependent on the efficacies of the scoring

functions employed and the availability of high-performance supercomputers (Ganesan &

Barakat, 2016).

In fact, although it was first developed to investigate molecular recognition between large and

small molecules, it is now also widely used to assist different tasks of drug discovery programs,

such as hit identification and optimization, drug repositioning, target identification, multi-target

ligand design, and repositioning as shown in Figure 2.7.

Figure ‎2.7: Main applications of molecular docking in current drug discovery (Pinzi, & Rastelli, 2019)

Page 60: Computational Approach Towards Exploring the

14

Moreover, docking allows understanding the relationships between different molecular targets

involved in a given disease, which is also of high relevance for polypharmacology and modern

drug discovery in general (Pinzi, & Rastelli, 2019).

7.1.7.2.4 Methods based on analysis of bioactivity spectra

Bioactivity spectra analyses methods work on the principle that compounds binding to same

target should display similar bioactivity spectra (i.e., the readouts from microarrays, cell lines

and in vitro screening). The bioactive spectra collected from different targets and assays are later

employed in the computational method to predict targets for the drugs. The important caveat of

this method is the need to perform expensive and time consuming experiments to collect

bioactivity spectra for different targets (Wang, et al., 2013; Kearnes and Pande, 2016).

3

4

5

6

7

8

9

Page 61: Computational Approach Towards Exploring the

12

10 Chapter Three

Methodology

3.1 Computational Method

Figure 3.1 shows the workflow of ROCS-based target fishing approach (RTFA) implanted in this

thesis. The work involves the following steps: the first was generation of active ligands database;

The second was generation of query compound; The third was similarity search and finally

molecular docking simulations.

Figure ‎10.1: Workflow of ROCS-based target fishing approach (RTFA)

3.1.1 Building 3D-ligands database

In order to cover the chemogenomic space, two databases were downloaded as SDF files: a) The

approved drug molecules (e-Drug3D) obtained from the Cheminformatic Tools and Databases

(contain 1822 molecular structures with a molecular weight ≤ 2000, last update: July 2016).

b) The co-crystallized ligands database obtained from Protein Data Bank (PDB) (www.rcsb.org)

including ligands that are used currently in PDB structures (contains 217000 entries). The two

databases were filtered using FILTER Software (Filter, 2013). In order to get rid of large

Page 62: Computational Approach Towards Exploring the

12

polypeptide drugs in the drug database, filtering was carried out using molecular weight as a

filter (180-800), for PDB database, the following parameters were adopted: allowed elements

(H, C, N, O, F, P, S, Cl, Br, I): number of heavy atoms (15-40); molecular weight (200-600);

number of ring system (0-5). After filtration, the drug database encompassed 1660 approved

drugs and the PDB had 6548 ligands. Subsequently, an ensemble of energetically accessible

conformers of the target representatives was generated using OMEGA software (OMEGA, 2013)

with default parameters.

3.1.2 ROCS similarity search

The ROCS run was carried out using vROCS which provides a single user interface from which

the user can build/edit ROCS queries, set up ROCS runs and visualize/analyze the results. There

are four primary tasks in vROCS available: a) Perform a simple ROCS run; b) Create a query

with a wizard; c) Create or edit a query manually; d) Perform a ROCS validation.

Queries from natural compounds were built in the vROCS query editor after exporting a line

notation describing the structure of chemical species (SMILES) and are automatically added to

this list for the current vROCS session. The query highlighted in blue is the selected (active)

query as shown in figure 3.2.

Figure ‎10.2: Simple run window display in vRocs user interface after generation of quercetin query.

Page 63: Computational Approach Towards Exploring the

11

A simple run was performed which aligns a database of precomputed molecular conformers

against natural queries. For each molecule in the chemogenomic database it overlays every

conformer based on molecular shape and chemical features. The conformers are scored based

upon the Gaussian overlap to the query and the best scoring conformer is reported. The adopted

score in our research was TanimotoCombo (shape + color). The molecules in the database are

finally ranked by the scores for their best aligned conformers. The higher TanimotoCombo score,

the better shape and chemical-feature match exists between molecules.

The ROCS run was carried out with the following parameters: Score to use for ranking the hit

list (rank by = TanimotoCombo); Maximum number of overlays returned for each comparison of

a database molecule with a query molecule; Keep a hit list with the highest score; Other

parameters as kept as default.

3.1.3 Docking simulations

3.1.3.1 Preparation of proteins

The 3D coordinates of target proteins had been downloaded from Protein Data Bank, (PDB;

https://www.rcsb.org) as seen in table 3.1, as (.pdb) files. Subsequently, hydrogen atoms were

added to proteins using Accelrys Discovery studio (DS) Visualizer.

Table ‎10.1: Proteins data obtained from Protein data bank.

Target PDB code Cpd.ID Origin References

CDK2 2DUV 371 Homo sapiens Lee, et al., 2007

CK2α 3AMY AGI Homo sapiens Kinoshita, et al.,

2013

DAPK1 5AV3 KMP Homo sapiens Yokoyama, et al.,

2015

DPP-4 3HAC 361 Homo sapiens Edmondson, et al.,

(2009).

PPAR-γ 3SZ1 LU2 Homo sapiens Puhl, et al., (2012).

PDK1 3NUN JMZ Homo sapiens Medina, et al., 2010

PIM1 4XH6 HUL Homo sapiens Chao, et al., 2015

GSK-3β 3Q3B 55E Homo sapiens Coffman, et al.,

(2011).

Tank 2 4HL5 15W Homo sapiens Narwal,et al., 2013

CDK2:Cycline dependent kinase2, CK2α: Casine kinase 2 alpha, Cpd.ID: Co-Crystallized ligand identification,

DAPK1: Death associated protein kinase1, DPP-4: Dipeptidyl peptidase, ERα: Estrogen receptor alpha, ERβ:

Esrtogen receptor beta, GSK-3β: Glycogen synthase kinase-3, PDB: Protein data bank, PDK1: Phosphoinositide-dependent kinase-1, PIM1: Proviral integration site for moloney murine leukaemia virus,

PPAR-γ: Peroxisome proliferator activated receptor gamma, PTP1B: Protein tyrosine phosphatase 1B, Tank-2:

Tankyrase 2

Page 64: Computational Approach Towards Exploring the

15

The target proteins binding sites were identified using pdb2receptor module within OEDocking

version 3.2.0.2 software suite (OEDOCKING, 2015). PDB2RECEPTOR is a utility program for

converting a protein-ligand complex into a receptor. It takes as an input the structure of the

protein-ligand complex and the name of a residue identifying a ligand bound to the active site.

The output receptor files were saved as OEBinary file (.oeb) files which are compatible with the

docking software.

3.1.3.2 Preparation of ligands

The chemical structure of natural products were sketched in Marvin Sketch (version 16.10.24)

and saved in molfile format. Afterwards, conformation space of compounds were explored by

generating energetically accessible conformers using OMEGA software (OMEGA 2.5.1.4, 2013)

and the generated conformers were saved in (.Sdf) format.

OMEGA is a conformation generator of molecules. OMEGA is composed of two main

components; model building and torsion driving. The software builds initial models of structures

by assembling fragment templates along sigma bonds. Once an initial model of a structure is

constructed, or given as input, OMEGA generates additional models by enumerating (a) ring

conformations, (b) invertible nitrogen atoms. Ring conformations are taken from the same

fragment library used to build an initial model, OMEGA detaches all exocyclic substituents from

a ring system, aligns and attaches them relative to the new ring conformation. OMEGA attempts

to generate every possible combination of ring conformations possible for a given structure. On

the other hand, nitrogens that have pyramidal geometry, no stereochemistry specified, no more

than one hydrogen, are three valent, and have no more than three ring-bonds are considered by

OMEGA to be invertible.

OMEGA begins the torsion search process by examining the molecular graph and determining

the bonds that may freely rotate. Exhaustive depth first torsion search is performed on each

fragment, and the resulting conformers are placed into a list sorted by energy. Entire structures

are assembled by combining the lowest energy set of fragments, and then the next lowest set,

until the search is terminated. The search will terminate when the limit on the total number of

conformers that may be generated is exceeded, the fragment list is exhausted, or the sum of the

fragment energies exceeds the energy window of the global minimum structure. The best

conformers identified in the torsion search are rank ordered by energy.

Page 65: Computational Approach Towards Exploring the

16

3.1.3.3 Docking

Natural compounds were docked into active site of target proteins using FRED software version

3.2.0.2 (FRED, 2015) within the OEDocking suite in the presence of the explicit water

molecules.

FRED is a fast rigid exhaustive protein-ligand docking program, which makes use of a pre-

generated multi-conformers database and a single receptor file as input and output molecules

most likely to bind to the receptor (FRED, 2015). The input files to this program is: receptor

saved as specialized OEBinary file (.oeb) files obtained from crystallographic structures of the

target proteins as shown in protein preparation section above; and one or more drug-like

molecules to be docked. The output is the docked pose of the molecules and information about

the dock score (Chemgauss4). The protein structures and ligands conformers were treated as

rigid units during docking process. The top scoring poses were optimized and assigned a final

score.

Here we could remember that the receptor is a specialized OEBinary file (i.e., .oeb or .oeb.gz

file) that contains the structure of a target protein, a negative image that describes the shape of

the active site and additional information about the location and characteristics of its binding

pocket, also the structure of a ligand bound to the active site and extra molecules that do not

affect either docking or scoring generally as water or other solvent molecules.

This docking engine takes a multiconformer database of one or more ligands, a target protein

receptor, that contains the structure of a target protein and additional information about the

location and characteristics of its binding pocket. The ligand conformers and protein structure

are treated as rigid during the docking process. FRED's docking strategy is to exhaustively score

all possible positions of each ligand in the active site (OEDOCKING, 2015). The exhaustive

search is based on rigid rotations and translations of each conformer. Therefore, it avoids

sampling issues associated with stochastic methods; semi random method.

Page 66: Computational Approach Towards Exploring the

17

11 Chapter Four

Results and Discussion

11.1 Background

Natural products contribute significantly to drug discovery research with a rich source of

compounds and provide inherently large-scale of structural diversity than synthetic compounds.

Modern drug discovery aims to identify hits that induce desired biological effects in cell culture

or animal models. Historically, nature has been an important source for such molecules,

however, the binding mechanisms of the identified molecules are often unknown, and

determining their underlying molecular targets has become an integral part of the drug discovery

process (Schenone, et al., 2013).

Current experimental target identification approaches can rarely achieve large-scale drug target

profiling (Verhelst & Bogyo, 2005). As a result, the development of in-silico drug target

profiling approaches that can effectively prioritize supposed on and off-targets for experimental

validation will be important for the success of current and future drug discovery programs.

In-silico target fishing methods can be classified as structure based or ligand based approaches

(Schenone, et al., 2013). Currently, ligand based approaches remain the standard for

computational target prediction, as this approach does not depend on the availability of protein

structures or prior experimental measurements. The rationale behind ligand based approaches is

the chemical similarity principle, which states that structurally similar compounds often share

similar bioactivities. To compare chemical similarity between compounds, each molecule is

encoded as a substructure fingerprint, and the degree of similarity is quantified by shared bits

using a Tanimoto index. To predict the drug targets for the query ligands, the compounds are

used to search the bioactivity databases, and putative drug targets are inferred from annotated

ligands in the database that share the highest chemical similarity to the query ligand (Lo, et al.,

2016).

Target fishing is considered as complementary process to experimental screening approaches as

it is not possible to test each compound against every possible target. This work is ligand-based

computational method which involve an efficient similarity measure and reliable scoring method

(Schenone, et al., 2013).

Page 67: Computational Approach Towards Exploring the

18

This study was designed to explore the polypharmacology of S. officinalis and U. dioica, well

known plants in our region, using ligand based target fishing approach and focusing on their

reported anticancer, antidiabetic and antihyperlipidemic bioactivities. Only thet op ranked targets

had been taken into consideration in the discussion below.

4.2 Polypharmacologyof S. officinalis

S. officinalis has been conventionally used for the treatment of various ailments since ancient

times at various parts of the world. Findings from in-vitro and several clinical studies supporting

the evidence of its medicinal uses such as cognitive, antioxidant, antimicrobial, anticancer,

antidementia, hypoglycemic, and hypolipidemic agents (Sharma & Schaefer, 2019).

The main phytochemical components of S. officinalis species are terpenoids and phenolic

components. The phenolic components can be divided into phenolic acids (caffeic, vanillic,

ferulic, and rosmarinic acids) and flavonoids (luteolin, apigenin, and quercetin) (Jakovljevic, et

al., 2019).

Terpenoids possess antitumor, anti-inflammatory, antibacterial, antiviral, antimalarial,

cardiovascular, antioxidant and hypoglycemic activities (Yang, et al., 2019). Flavonoids are

shown to have antioxidant activity, free radical scavenging capacity, coronary heart disease

prevention, hepatoprotective, anti-inflammatory, and anticancer activities, while some flavonoids

exhibit potential antiviral activities (Kumar & Pandey, 2013).

4.2.1 Reported anticancer, antidiabetic and hypolipidemic targets of S. officinalis

Several suggested mechanisms for S. officinalis anticancer effects were reported in the literature.

The suggested mechanisms for the anticancer role of dietary intake of S. officinalis include the

interaction with signaling pathways, such as estrogen receptor (ER)-mediated, Mitogen-

Activated Protein Kinase/extracellular signal-regulated kinase (MAPK/ERK), nuclear

transcription factor-kappa B (NF-kB), Vascular endothelial growth factor/ Vascular endothelial

growth factor receptor (VEGF/VEGFR), phosphatase and tensin homolog/ phosphatidylinositol

3-kinase /Protein Kinase B (PTEN/PI3K/Akt), p53, and mitochondria-dependent pathways

(Xavier, et al., 2009; Jian et al., 2016).

Recent pharmacological investigations demonstrated that different extracts of aerial parts of S.

officinalis are able to decrease blood glucose in normal and diabetic conditions. The mechanisms

suggested for hypoglycemic effect of S. officinalis include an inhibition of hepatocyte

gluconeogenesis and decrease of insulin resistance through stimulation of peroxisome

Page 68: Computational Approach Towards Exploring the

19

proliferator activated receptor-γ (PPAR-γ) (Christensen, et al., 2010; Ghorbani & Esmaeilizadeh,

2017).

The possible mechanism of hypolipidemic activity of S. officinalis may be due to free radical-

scavenging and antioxidant activities which might be attributed to the presence of flavonoids. In

addition, the decrease in the level of cholesterol may be due to the activity of Cholesterol 7-α

hydroxylase which works to lower blood cholesterol level by converting it to bile salts (Uthandi

& Karuppasamy, 2012; Abdulhussein, et al., 2019)

Using RTFA, we had discovered that many of S. officinalis natural constituents could bind to

ER, MAPK14, PIK3CG and PPAR-γ such as apigenin, luteolin, oleanolic acid and quercetin.

As shown in table 4.1, four on-targets and twelve off-targets related to anticancer activity of S.

officinalis, only one on-target and four off-targets related to antidiabetic activity of S. officinalis,

in addition to one off-target related to hypolipidemic activity of S. officinalis, were identified by

the adopted RTFA protocol.

Table ‎11.1: On-targets and off-targets of S. officinalis

Anticancer Targets Antidiabetic Targets Hypolipidemic Targets

On-Target Off-Targets On-Targets Off-Targets On-Targets Off-Targets

1 ERα CK2 PPAR-γ GP PTP1B

2 ERβ PIM1 DPP-4

3 MAPK14 DAPK1 PTP1B

4 PIK3CG PDK1 PDK1

5 Tank2

6 GLO-I

7 FGFR1

8 RPTP-γ

9 CDK2

10 17β-HSD1

11 HSP90-α

12 Mcl-1

17β-HSD: 17 Beta Hydroxysteriod dehydrogenase 1,CDK2: Cycline dependent kinase 2, CK2: Casine kinase 2,

DAPK1: Death associated protein kinase1, DPP-4: Dipeptidyl peptidase-4, ERα: Estrogen receptor alpha,

ERβ:Estrogen receptor beta, FGFR1: Fibroblast growth factor receptor 1, GLO-1: Glyoxalase 1, GP: Glycogen

phosphorylase, HSP90-α: Heat shock protein 90 alpha, MAPK14: Mitogen activated protein kinase 14, Mcl-1:

myloid cell leukemia 1, PDK1: phosphoinositide-dependent kinase1, PIK3CG: phosphatidylinositol-4,5-

bisphosphat 3-kinase catalytic subunit gamma, PIM1: proviral integration site for moloney murine leukaemia

virus, PPAR-γ: peroxisome proliferator activated receptor gamma, PTP1B: protein tyrosine phosphatase 1B,

RPTP-γ: Receptor type protein tyrosine phosphatase gamma, Tank2: Tankyrase2.

On-Targets: Known targets, reported in the literature, for the specified bioactivity of the query compound.

Off-Targets:Unknown targets, not reported in the literature, for the specified bioactivity of the query compound.

Page 69: Computational Approach Towards Exploring the

51

Analysis of the results that retrieved from RTFA highlights the importance of this approach in

clarifying the polypharmacology of S. officinalis especially the anticancer, antidiabetic and

hypolipidemic activities. RTFA clarify the ability of S. officinalis to bind with reported

anticancer targets (on-targets) as MAPK14, ERα, ERβ and PIK3CG as well as to bind with the

reported antidiabetic target such as PPAR-γ. Moreover, we had discovered many off-targets for

cancer, diabetes and hyperlipidemia that had been caught by S. officinalis constituents as CK2,

PIM1, PTP1B, PDK1…etc.

4.2.2 Targets identified using S. officinalis constituents as queries in RTFA

4.2.2.1 Apigenin

Apigenin, known chemically as 4′,5,7-trihydroxyflavone (as shown in table 2.2), belongs to the

flavone subclass and is abundant in a variety of vegetables, fruits and medicinal plants. It has

several reported biological activities, including antioxidant, anti-inflammatory, antitumor,

antidiabetic, neuroprotective (thus beneficial in amnesia and Alzheimer‘s disease) and

cardioprotective effects (Salehi et al., 2019).

Using RTFA and apigenin as query template, several targets related to cancer and diabetes were

identified, where all of them were on-targets : casein kinase II alpha (CK2α), proviral integration

site for moloney murine leukaemia virus 1 (PIM1), Death-associated protein kinase 1 (DAPK1),

Tankyrase 2 (Tank-2), mouse Glyoxalase-1 (GLo-1), Estrogen receptor α (ERα), Estrogen

receptor β (ERβ) and Glycogen phosphorylase (GP) as shown in table 4.2. This indicates the

biological importance of apigenin and the research interest in its multiple biological activities.

The first target, with the highest rank value was CK2α as shown in table 4.2. It is an ubiquitous,

pleiotropic, highly conserved and constitutively active serine/threonine kinase (Litchfield, 2003).

In many reports, the implication of CK2α in several pathologies is described, like neurodege-

nerative diseases (Parkinson‘s and Alzheimer‘s disease), inflammation, viral and parasite

infections as wellas cancer (Baier, et al., 2018).

CK2α enhances cancer phenotype by blocking apoptosis and simultaneously stimulating cell

growth. A number of cancers are associated with hyper activation and overexpression of CK2α

including breast, lung, prostate, colorectal, renal, and hematological malignancies (Ortega, et al.,

2014), thus CK2α could be considered as a promising target for cancer treatment.

Page 70: Computational Approach Towards Exploring the

54

CX-4945 from Cylene Pharmaceuticals, also known as Silmitasertib, is the first inhibitor of CK2

that has been qualified for human clinical trials (Siddiqui-Jain, et al., 2010). It has successfully

completed phase I, and is currently in phase II for cholangiocarcinoma treatment, which granted

it an Orphan Drug status for cholangiocarcinoma by Food and Drug Administration (FDA) in the

USA in January 2017 (Masłyk, et al., 2017). CX-4945 is an ATP- competitive inhibitor of

protein kinase CK2 and a highly selective, orally administered small molecule studied in

different types of human cancer research (Siddiqui-Jain, et al., 2010). Human clinical

characterization of CX-4945 as a single agent in solid tumors and multiple myeloma has shown

its promising pharmacokinetic, pharmacodynamics, and safety profiles (Padgett, et al., 2010). In

addition, CX-4945; when used in anticancer therapy, it may simultaneously prevent cancer-

associated candidiasis (Masłyk, et al., 2017 ).

In addition, In July 2020, Silmitasertib was granted Rare Pediatric Disease Designation in

medulloblastoma by the US FDA. Moreover, an Emergency Investigational New Drug (EIND)

was granted by the US FDA on August 27, 2020, for use of Silmitasertib in the treatment of

sever coronavirus disease of 2019 (COVID-19). Silmitasertib has great potential as a therapeutic

for COVID-19 and as it targets the host protein kinase CK2 pathway, virus mutations are

unlikely to affect either antiviral or anti-inflammatory efficacy of Silmitasertib (John, 2020).

Table ‎11.2: Pharmacological profiling for apigenin using ROCS

Target Disease Tanimoto coefficient Ref.

CK2 α a Cancer 2.000

Lolli, et al.,2012 & Liu,

et al., 2015

PIM1 a Cancer 1.860

Gadewal and Varma.

2012

DAPK1 a Cancer 1.824 Yokoyama et al.,2015

Tank2 a Cancer 1.686 Narwal et al., 2013

mGLO-I a Cancer 1.651 Zhang et al.,2016

ERα b Cancer 1.523 Maruthanila, et al., 2019

ERβ b

Cancer 1.436 Powers & Setzer. 2015

GP b T2DM 1.261 Paramaguru, et al., 2014

CK2α: Casein kinase two alpha, DAPK1: Death-associated protein kinase 1, ERα: Estrogen Receptor alpha, ERβ: Estrogen Receptor beta, GP: Glycogen Phospholylase, mGLo-1: mouse Glyoxalase, PIM1: Proviral

integration site for moloney murine leukaemia virus 1, Tank2: Tankyrase2.

a Has in-vitro and/or in-vivo evidence, b Has in-silico evidence.

Herein, the RTFA had identified CK2α as an anticancer target and this finding was supported by

Lolli et al., who assayed a panel of 16 flavonoids and related compounds for their ability to

Page 71: Computational Approach Towards Exploring the

52

inhibit CK2α. Among the tested compounds, apigenin had an inhibition activity with IC50 value

of 0.80 μM (Lolli, et al.,2012). In addition, we found that apigenin had a tanimoto coeff. of 2

which means apigenin itself was the ligand for CK2α.

The second on-target was PIM1 from the proto-oncogene family (This family includes three PIM

genes PIM1, PIM2 and PIM3), represents a novel class of constitutively active serine/threonine

kinases. PIM1 had been shown to play a pivotal role in cell survival, proliferation, and

differentiation (Wang, et al., 2001). In addition, PIM1 has been implicated in avoidance of

apoptosis (Rebello, et al., 2018) by interacting with other tumorigenic pathways, such as the

PI3K /AKT pathway (Warfel & Kraft, 2015). Moreover, PIM up-regulation can cause resistance

to conventional chemotherapy, radiotherapy and other therapeutics (Zemskova, et al., 2008). In

humans, the PIM1 oncogene is expressed in lymphoid and haematopoietic cells, (Kumar, et al.,

2005; Meeker, et al., 1990), prostate malignancies (Valdman, et al., 2004), squamous cell

carcinomas of the head and neck region (Jeon, et al., 2004), gastric carcinomas (Sepulveda, et

al., 2002) and colorectal carcinomas (Xu, et al., 2011). An investigation found that chromosomal

translocation of PIM1 leading to overexpression of PIM1 is involved in diffuse large cell

lymphoma, the most common form of non-Hodgkin's lymphoma (Akasaka, et al.,2000).Thus, the

strong connection between overexpression and mutation of PIM1 and cancer suggests that PIM1

kinase inhibition is a promising drug target in the treatment of cancer.

To date, most efforts to inhibit PIM in cancer treatment have focused on the use of ATP-

competitive drugs that target the kinase action of the protein, preventing it from phosphorylat ing

its downstream effectors (Wang & Sun, 2019). A number of small molecule PIM inhibitors have

been developed and some have progressed to clinical stages such as flavonoid inhibitor, SGI-

1776 (Yang, et al., 2013) and AZD1208 (Keeton, et al., 2012). They can be classified as the first

generation inhibitor (SGI-1776) and the next generation inhibitor (AZD1208) (Mondello, et al.,

2014).

Studies demonstrated that SGI-1776, the first generation imidazopyridazine-based Pim kinase

inhibitor to enter clinical study, exhibited potent antitumor activity in vivo as well as in vitro.

SGI-1776 also showed to sensitize drug resistant cells to cancer chemotherapy drugs by

inhibiting p-glycoprotein (Pgp)-mediated efflux and inducing apoptosis (Burger, et al., 2008).

SGI-1776 was evaluated in a Phase I clinical trials, but failed to progress, due to its

cardiotoxicity (Le, et al., 2015).

The second generation inhibitors were quickly followed, aiming to remove cardiotoxicity while

maintaining the PIM potency. Thus, SGI-9481 (also known as TP-3654) was developed which is

Page 72: Computational Approach Towards Exploring the

52

a pyrazolopyrimidine-based PIM inhibitor with improved potency and decreased cardiotoxicity

compared to thiazolidinedione-based inhibitor. Notably, this drug selectively targets PIM1, with

a Ki (inhibition constant) of 5 nm (Luszczak, et al., 2020).

PIM kinase inhibitors in the clinical studies such as AZD1208 (Thiazolidinedione-based),

CXR1002 and LGH447 have been shown to induce apoptosis affecting cell proliferation and

migration, high lighting the potential for targeting PIM as an anticancer therapy (Pierre, et al.,

2011; Keeton, et al., 2012; Yadav, et al., 2019).

Gadewal and Varma, investigate the binding of PIM1 and apigenin and found that apigenin

exhibited PIM1 kinase inhibitory activity with IC50 value of 0.94µM (Gadewal & Varma, 2012),

this support our RTFA findings which identified PIM1 as a potential anticancer target for

apigenin and the whole plant.

The third on-target was DAPK1. It is a 160-kDa cytoskeletal associated protein kinase that

consists of 1430 residues and belongs to the superfamily of calcium/calmodulin (Ca+2

/CaM)

regulated serine/threonine protein kinases (Shiloh, R., et al., 2014). DAPK1 was identified as a

mediator of γ-interferon-induced cell death and a tumor suppressor. It is also linked to activation

of autophagy (Zalckvar, et al., 2009). In addition, knockdown of DAPK1 expression induced

TRAIL-mediated apoptosis in human endometrial adenocarcinoma cells, suggesting that DAPK1

may be a candidate molecule for advanced endometrial adenocarcinomas (Bai, et al., 2010).

Moreover, DAPK1 is an upstream negative regulator of peptidylprolyl cis/trans isomerase, never

in mitosis A(NIMA)-interacting 1 (Pin1) that activates numerous oncogenes and suppresses

many tumor suppressors, suggesting that activating DAPK1 effectively inhibits multiple

oncogenic signaling pathways (Huang, et al., 2014). It has also been reported that DAPK1 might

be oncogenic on certain cellular condition such as p53 mutant cancers (Zhao, et al., 2015).

Therefore, more studies of DAPK1 regulatory mechanisms and specific DAPK1 activators are

needed.

Yokoyama, et al., investigated the binding affinities of 17 natural flavonoids to DAPK1 and

explained their different affinities by means of the crystallographic analysis of DAPK1 with

selected flavonoids. The binding affinity of apigenin to DAPK1 had IC50 of 31 ± 3.6 µM

(Yokoyama, et al., 2015) and this go with our results from RTFA which identified DAPK 1 as a

possible target that might contribute to the anticancer activity of our plant.

Page 73: Computational Approach Towards Exploring the

51

The fourth on-target of apigenin was Tank2. Tank2 is a multifunctional poly ADP(adenisine

diphosphate)-ribose polymerase (PARP) that regulates a variety of cellular processes, including

telomere maintenance (allows continued proliferation), oncogenic pathways, mitosis and DNA

repair and cell death. Therefore, Tank2 inhibitors may be effective targets for cancer treatment

(Haikarainen, et al., 2014).

Numerous studies have reported the importance and utility of tankyrase inhibitors as cancer

therapeutics (Quackenbush, et al., 2016). Consequently, a number of tankyrase inhibitors with

promising therapeutic effects have been developed, including XAV939 (Huang, et al., 2009;

Bao, et al., 2012; Busch, et al., 2013;), IWR-1, G007-LK (Lau, et al., 2013), JW55 (Waaler, et

al., 2012), AZ1366 (Quackenbush, et al., 2016), JW 74 (Tian, et al., 2013; Stratford, et al., 2014)

and NVP-TNKS656 (Arqués, et al., 2016; Wang et al, 2016).

Narwal, et al., had performed a systematic screening of Tank2 inhibitory activity using 500

natural and naturally derived flavonoids and found that apigenin is Tank2 inhibitor with IC50 of

2.9 μM (Narwal, et al., 2013).

The fifth on-target was Glyoxalase I (GLO-I). GLO-I is the first and rate-limiting zinc enzyme in

the mammalian glyoxalase system for catalyzing the conversion of toxic α-oxoaldehydes to

nontoxic α-hydroxacids (Sousa, et al., 2103). The glyoxalase pathway is an antioxidant defense

mechanism. GLO-I has been reported to be frequently overexpressed in various types of cancer

cells, and has been expected as an attractive target for the development of new anticancer drugs

(Thornalley & Rabbani, 2011).

A novel inhibitor of human GLO-I, named TLSC702, was discovered by in silico screening

method with IC50 value of 2.0 µM. TLSC702 inhibits the proliferation of human leukemia and

lung cancer cells and induces apoptosis in a dose-dependent manner in both cancer cells. Taken

together, TLSC702 could become a unique seed compound for the generation of novel

chemotherapeutic drugs targeting GLO-I dependent human tumors (Takasawa, et al., 2016).

Apigenin inhibits the activity of GLO-1 and had IC50 value of 70 µM (Zhang, et al., 2016).

The sixth on-target was ERα, which is a member of the nuclear receptor superfamily of

transcription factors (Jameera, et al., 2107). It plays a vital role in the delineation and

maintenance of neural, skeletal, cardiovascular, and reproductive tissues (Hsu, et al., 2017).

However, ERα is considered as an oncogene and is mainly responsible for the breast cancer

initiation and progression (Folkerd, et al., 2010). Currently, compounds which effectively

Page 74: Computational Approach Towards Exploring the

55

modify ERα transcriptional activity are found beneficial in the treatment of osteoporosis,

cardiovascular disease, and breast cancer (Jordan, 2007; Hua, et al., 2018).

Herein, the RTFA had presented ERα as anticancer target and this supported by Maruthanila, et

al. In their study, they had screened selected natural ligands and their binding features with ERα

and found that apigenin has a strong binding to ERα with Glide Scores of -8.92 Kcal mol-1

using

molecular docking simulations (Maruthanila et al., 2019).

The seventh on-target was ERβ, which is a members of the nuclear receptor superfamily of

transcription factors. Estrogen is a steroid hormone that has critical roles in reproductive

development, bone homeostasis, cardiovascular remodeling and brain functions. However,

estrogen also promotes mammary, ovarian and endometrial tumorigenesis. ERβ is considered as

a tumor suppressor (Hua, et al., 2018) and its activation is an important mechanism for cancer

prevention (Yang, et al., 2015).

Powers & Setzer (2015) studded the binding of apigenin and ERβ and found that the molecular

docking energy of apigenin with ERβ had a value of −97.3 KJ/mol.

These findings could increase confidence in our approach (RTFA) for exploring the mechanisms

of the anticancer activity of S. officinalis.

The last on target was glycogen phosphorylase (GP) as shown in table 4.2. GP is a large protein

composed of two subunits (α and β) and present in muscles (m GP), brain (b GP) and liver

(l GP). GP exists under different conformational states symbolizing its activation degree (α

subunit is the phosphorylated and active unit while β subunit is the dephosphorylated and

inactive unit) (Gaboriaud & Skaltsounis, 2013). GP catalyzes the breakdown of glycogen and

largely contributes to hepatic glucose production. This breakdown of glycogen is enhanced

during fasting and reduced by GP inhibition. Moreover, GP inhibition enhances glycogen build

up in skeletal muscle and enhances hepatic glucose uptake that contributes to glucose clearance

from blood. These effects together makes GP inhibition an attractive target to modulate glucose

levels in diabetes (Nagy, et al., 2013; Stravodimos, et al., 2018).

Numerous GP inhibitors have been published in the last two decades. Indeed, a novel GP

inhibitor, Ingliforib in clinical study was able to reduce glucagon-induced hyperglycemia (Agius,

2007). Also it markedly reduces myocardial ischemic injury in vitro and in vivo; this may

represent a viable approach for both achieving clinical cardio protection and treating diabetic

patients at increased risk of cardiovascular disease (Tracey, et al., 2004).

Page 75: Computational Approach Towards Exploring the

56

KB228 is another novel and potent GP inhibitor was tested in vitro and in vivo under

normoglycemic and diabetic conditions. It reduced serum glucose levels and increased hepatic

glycogen content under both normoglyce-mic and insulin resistant, hyperglycemic conditions

(Nagy, et al., 2013).

Moreover, several GP inhibitors were tested in clinical trials and passing phase I, confirming the

safety of these drugs. Namely, CP-316819 (Pfizer), AVE56588 (Sanofi-Aventis) and

GSK1362885 (GlaxoSmithKline) completed phase I, while CP-368296 (Ingliforib, Pfizer) and

PSN-357(Prosidion, now Astellas Pharma) advanced to phase II. However, unfortunately, the

clinical studies on GP inhibitors were halted after phase II, but the reasons were not

communicated (Nagy,et al., 2018).

Herein, GP was identified as a potential target and this finding was supported by molecular

docking simulations study carried out by Paramaguruet, et al., 2014. They had investigated the

potential interaction of apigenin with GP using Autodock 4.2. and they had reported that

apigenin could bind with GP making three hydrogen bond interactions with the active site

residues Glu88, His377 and Asn484. The estimated free energy of binding of apigenin was found

to be −8.08 Kcal mol−1

with estimated inhibition constant (Ki) of 1.2 µM (Paramaguruet, et al.,

2014). These findings could be responsible, even partially, for its beneficial antidiabetic effects.

4.2.2.2 Carnosol

Carnosol is an ortho-diphenolic diterpene (as seen in table 2.3), naturally occurring compound of

the labiate herbs rosemary and sage. It has multiple beneficial medicinal effects including anti-

oxidante, anti-inflammatory, antimicrobial, antidiabetic and anticancer in various disease

models. (Johnson, 2011; Farkhondeh, et al., 2016).

By the application of RTFA on carnosol as query template, four targets related to cancer, and

diabetes were identified, that were off-targets as shown in table 4.3.

The first potential off-target was the human fibroblast growth factor receptor 1 (FGFR1),which

is a member of the FGFRs family that consists of four members (FGFR 1- 4). FGFRs belong to

the family of the receptor tyrosine kinases (RTKs) and play fundamental roles in several basic

biological and physiological processes by participating in the regulation of cell proliferation,

migration, differentiation, survival, apoptosis, metabolism, and angiogenesis (Carter, et al., 2015;

Ornitz & Itoh, 2015). Over activation of FGFR1 signaling occurs in many types of cancer due to

gene amplification, mutations or translocations (Wang, et al., 2014). FGFR1 is expressed in a

Page 76: Computational Approach Towards Exploring the

57

wide variety of cell types and tissues, including many malignancies such as breast, lung,

mammary gland, pancreatic and prostate cancers as well as the advanced stage of head and neck

squamous cell carcinoma. Moreover, the inhibition of FGFR-1 resulted in significant growth

inhibition in pancreatic, breast, prostate, lung and several other cancer cell lines (Navid, et al.,

2011). On this basis, FGFR has been validated as an attractive target for targeted cancer therapy

(Hallinan, et al., 2016; Giuseppe, et al., 2019).

Table ‎11.3: Pharmacological profiling for carnosol using ROCS

Target Disease Tanimoto coefficient Docking score

FGFR1 Cancer 1.081 -9.337

Tank2 Cancer 1.079 -11.561

CK2 Cancer 1.061 -08.344

DPP-4 T2DM 1.022 -07.457

CK2: Casine kinase 2, DPP-4: Dipeptidyl peptidase-4, FGFR1: Fibroblast growth factor receptor1, Tank2:

Tankyrase2.

In recent years, several small molecule FGFR inhibitors have been reported, and some of them

are now in clinical trials. The early examples of FGFR inhibitors are predominantly multi-

targeting drugs, such as nintedanib, lenvatinib, dovitinib, and lucitanib. Currently, several FGFR-

selective inhibitors have progressed into clinical trials including JNJ-42756493, AZD4547 and

NVP-BGJ398 (Zhang, et al., 2016).

Erdafitinib (Balversa™, Janssen Pharmaceutical Companies) is a pan-fibroblast growth factor

receptor (FGFR) inhibitor that was recently approved in the USA for the treatment of locally

advanced or metastatic urothelial carcinoma. The drug is also being investigated as a treatment

for other cancers including cholangiocarcinoma, liver cancer, non-small cell lung cancer,

prostate cancer, lymphoma and oesophageal cancer (Markham, 2019).

Herein, RTFA has identified carnosol as potential inhibitor of FGFR1. For further invistigation,

docking simulations using molecular docking has revealed the ability of carnosol to bind to the

FGFR1 target with good docking score (-9.34).

The obtained results highlight the possibility of FGFR1 to be a potential target and be partially

responsible for the anticancer activity of our plant and add in-silico evidence for the reported

polypharmacology of natural products.

Furthermore, the obtained data suggested that carnosol and apigenin, two different natural

compounds from S. officinalis, could have activity against the same targets (Tank2 and CK2) and

Page 77: Computational Approach Towards Exploring the

58

this support the fact that the biological activity of plants (herein, anticancer activity) could be

resulted from synergistic action of its multiple constituents on the same target or on different

targets related to pathogenesis of the same diseases as shown here.

The fourth off-target was the dipeptidyl peptidase-4 (DPP-4) which is a member of the prolyl

oligopeptidase family of related proteins and is one of the newest targets for T2DM treatment

(Deacon, et al., 2019).

DPP‐4 inhibitors are effective antihyperglycaemic agents by virtue of their ability to

inhibit the breakdown of the active form of the incretin hormones glucagon‐like

peptide1 (GLP‐1) and glucose‐dependent insulinotropic peptide (GIP) where both

hormones have very short half-lives (approximately 2 min) (Yang, et al., 2014). This

results in increased plasma levels of the intact and, thus, biologically active form of

both incretin hormones, which results in an improvement of glycaemic control

primarily via augmentation of glucose‐stimulated insulin secretion and inhibition of

glucagon release by the beta and alpha cells of the pancreas, respectively as shown in

figure 4.1 (Deacon, et al., 2019).

Figure ‎11.1: Mechanism of action for GIP, GLP-1 analogues and DPP4 inhibitors in controlling T2DM

(Deacon, et al., 2019)

Page 78: Computational Approach Towards Exploring the

59

Several structurally diverse DPP‐4 inhibitors have become established therapeutically, the most

widespread of which are sitagliptin, vildagliptin, saxagliptin, linagliptin and alogliptin. Presently,

no less than eleven DPP‐4 inhibitors have been approved by regulatory authorities worldwide,

although some with more limited geographical availability (Deacon, et al., 2019).

The validity of the captured four proteins as potential targets for carnosol was evaluated by

molecular docking simulation according to our adopted experimental procedures and the

obtained results were shown in table 4.3. These findings could promote further studies on these

new off-targets which could give rise to the discovery of new anticancer and antidiabetic agents.

Among these off-targets, Tank2 had the highest docking score and this attract us to further

investigate its binding with carnisol at molecular level as seen in figure 4.2.

A B

Figure ‎11.2: A : Detailed view of docked carnosol and the corresponding interacting amino acid within the

binding site of Tank2, B : Detailed view of co-crystallized structure (G9W, PDB code: 5C5Q) and the

corresponding interacting amino acid within the binding site of Tank2 (Green lines refere to hydrogen bonds

and black lines refere to hydrophobic interactions)

As shown in figure 4.2, a strong network of hydrogen bonds between hydroxyl groups and keto

group of carnosol and key amino acids in the binding site of Tank2 including ILE1075,

GLY1032, and SER1068 similar to the co-crystallized structure. In addition to the aromatic rings

which occupy a lipophilic cavity composed of TYR1060, TRY1050 and TYR1071. These

interactions indicated a valid and substantial a strong binding with Tank2 and support our

postulation that carnosol could contribute to anticancer effect of S. officinalis and it is a good

candidate for further in-vitro / in-vivo experimental validation as Tank2 inhibitor.

Page 79: Computational Approach Towards Exploring the

61

4.2.2.3 Cirsimaritin

Cirsimaritin also known as 4',5-Dihydroxy-6,7-dimethoxyflavone (as shown in table 2.2),

belongs to the class of organic compounds known as 7-O-methylated flavonoids. It is an active

flavone associated with several potent pharmacological effects including antioxidant, antiinfl-

ammatory, antimicrobial, antidiabetic, anticancer, antagonistic properties, neurological effects,

cardiovascular, and hepatoprotective (Mahmood & Alkhathlan. 2019).

As we had applied our adopted approach on cirsimaritin as query template, several targets related

to cancer and diabetes were identified, where all of them were off-targets as shown in table 4.4.

Table ‎11.4: Pharmacological profiling for Cirsimaritin using ROCS

Target Disease Tanimoto Coefficient Docking Score

PIM1 Cancer 1.870 -11.538

CK2 α Cancer 1.760 -12.627

mGLO-1 Cancer 1.635 -02.491

PPAR-γ T2DM 1.624 -10.582

DAPK1 Cancer 1.623 -11.661

Tank2 Cancer 1.472 -13.191

ERα Cancer 1.366 -14.323

ERβ Cancer 1.365 -13.639

CK2α: Casein kinase two alpha, DAPK1: Death-associated protein kinase 1, ERα: Estrogen Receptor alpha,

ERβ: Estrogen Receptor beta, GP: Glycogen Phospholylase, mGLo-1: Glyoxalase 1, PPAR-γ: Peroxisome

proliferator activated receptor gamma, PIM1: Proviral integration site for moloney murine leukaemia virus 1,

Tank2: Tankyrase 2.

In-silico target fishing had led to identification of eight new off-targets for cirsimaritin: PIM1,

CK2α, GLO-1, PPAR-γ, DAPK1, Tank2, ERα and ERβ. All of these targets were discussed in

section 4.2.2.1.1 except PPAR-γ.

PPAR-γ is a member of the nuclear hormone receptor family of ligand-activated transcription

factor. It is highly presents in adipose tissues and plays a key role in regulating the insulin

sensitivity, adipocyte differentiation, inflammation and cell growth. PPAR-γ is generally

regarded as a molecular target for the thiazolidinedione class of antidiabetic drugs, as it plays a

key role in the generation and development of diabetes. Recent studies have shown that PPAR-γ

agonists, including rosiglitazone and pioglitazone, may be used as insulin sensitizers in target

tissues to lower glucose, as well as fatty acid levels in T2DM patients (Jian, et al., 2018).

Page 80: Computational Approach Towards Exploring the

64

The validity of these proteins as potential targets for cirsimaritin was evaluated by molecular

docking simulation according to our adopted experimental procedures and the obtained results

were shown in table 4.4.

Here we had chosen PPAR-γ to show its docking image with cirsimaritin at molecular level as

seen in figure 4.3, where a potential hydrogen bonding between cirsimaritin and key amino acids

at the binding site of PPAR-γ including ILE281, HIS266 and LYS265. In addition, the aromatic

rings occupy a lipophilic cavity composed of ILE341 and CYS285.

Figure ‎11.3: Detailed view of docked cirsimaritin and the corresponding interacting amino acids within the

binding site of PPAR-γ (Green lines refere to hydrogen bonds and black lines refere to hydrophobic

interactions).

4.2.2.4 Corosolic Acid

Corosolic acid is a pentacyclic triterpene, also known as 2 alpha-hydroxy ursolic acid or glucosol

(as seen in table 2.3), discovered in numerous medicinal herb (Baba, et al., 2018). It is reported

to exhibit antidiabetic, antihyperlipidemic, antioxidant, antiinflammatory, antiproliferative,

antifungal, antiviral, antineoplastic, osteoblastic and protein kinase C inhibition activity (Miura,

et al., 2012; Wang, et al., 2020).

To identify the possible anticancer, antidiabetic and hypolipidemic targets for corosolic acid,

RTFA had been used and several targets had been identified where all of them were off-targets

as shown in table 4.5.

Page 81: Computational Approach Towards Exploring the

62

The first off-target identified was the human receptor type protein tyrosin phosphatase gamma

(RPTP-γ). It is a member of the protein tyrosine phosphatase (PTP) family. PTPs are known to

be signaling molecules that regulate a variety of cellular processes including cell growth,

differentiation, mitotic cycle and oncogenic transformation.This PTP possesses an extracellular

region, a single trans-membrane region, and two intra-cytoplasmic catalytic domains, and thus

represents a receptor-type PTP. RPTP is located in a chromosomal region that is frequently

deleted in renal cell carcinoma and lung carcinoma, thus is thought to be a candidate tumor

suppressor gene (Xu & Fisher, 2012).

Table ‎11.5: Pharmacological profiling for corosolic acid using ROCS

Target Disease Tanimoto Coefficient Docking score

RPTP-γ Cancer 0.993 -7.695

Tank 2 Cancer 0.985 -8.949

CDK2 Cancer 0.957 -6.233

PPAR-γ T2DM 0.945 -5.460

ERα Cancer 0.938 -11.21

CDK2: Cyclin dependent, ERα : Estrogen receptor alpha , PPAR-γ: Peroxisome proliferator activated receptor

gamma, RPTP-γ :Receptor type protein tyrosine phosphatase gamma,Tank2: Tankyrase 2.

Tank2 and ERα were discussed before in section 4.2.2.1, while the fourth target, PPAR-γ, was

discussed in section 4.2.2.3.

The third target was the human cyclin dependent kinases 2 (CDK2) which is a member of CDKs

family. CDKs are serine/threonine protein kinases. This family has critical functions in cell cycle

regulation and controlling of transcriptional elongation. Moreover, dysregulated CDKs have

been linked to cancer initiation and progression. Pharmacological CDK inhibition has recently

emerged as a novel and promising approach in cancer therapy (García-Reyes, et al., 2018).

Two CDK inhibitors have been approved for marketing. Palbociclib developed by Pfizer is the

first CDK inhibitor approved to enter the market in February 2015 for the treatment of metastatic

breast cancer (Gupta, et al., (2016). Ribociclib developed by Novartis is the second CDK

inhibitor approved by FDA in 2017 for the treatment of advanced breast cancer in combination

with an aromatase inhibitor (Zhao,et al., 2019)

In-silico molecular docking simulations were conducted to validate the possibility of binding of

corosolic acid with its new off-targets. The obtained results clarified that the corosolic acid was

Page 82: Computational Approach Towards Exploring the

62

successfully docked within the active site of RPTP-γ, Tank2, CDK2, PPAR-γ and ERα with the

relatively good scores as shown in table 4.5.

4.2.2.5 Ellagic acid

Ellagic acid (EA), a fused four ring compound ( as shown in table 2.2), is a naturally occurring

tannic acid derivative that is widely found in fruits, vegetables and other foods. Many studies

have shown that EA possesses strong antioxidant, anti-inflammatory, antiproliferative effects,

apoptosis induction, metastasis inhibition and anticarcinogenic properties, suggesting the strong

anticancer activity of ellagic acid (Cozza, et al., 2006; Sepúlveda,et al., 2012).

To identify the possible anticancer, antidiabetic and hyperlipidemic targets for EA, RTFA had

been used. Numerous targets had been identified, some of them were on-targets as CK2, ERα

and CDK2, while others were off-targets as shown in table 4.6.

4.2.2.5.1 On-targets of Ellagic acid

The first on-target was CK2. EA inhibitory capacity for CK2 had been reported with IC50 value

of 0.04 µM (Alchab, et al., 2015), as it considered as a potent inhibitor of CK2.

Table ‎11.6: Pharmacological profiling for ellagic Acid using ROCS

Target Disease Tanimoto

Coefficient DockingScore Ref.

CK2 a Cancer 1.188 Alchab, et al.,

2015

mGLO1 Cancer 1.173 -02.079

Tank2 Cancer 1.172 -12.392

PIM1 Cancer 1.133 -11.940

PDK1 Cancer & T2DM 1.127 -12.660

17β-HSD1 Cancer 1.108 -14.429

ERα a Cancer 1.092 Pang, et al., 2018.

CDK2 b Cancer 1.069

Mohan, & Latha,

2018

17β-HSD1: 17 Beta Hydroxysteroiddehydrogenase type1, CK2: Casine kinase 2, CDK2: Cycline dependent

kinase2, ERα: Estrogen Receptor alpha, mGLO1: mouse Glyoxalase I, PDK1: Phosphoinositide-dependent

protein kinase-1, PIM1: Proviral integration site for moloney murine leukaemia virus 1, Tank2: Tankyrase 2.

a: Has in-vitro or in-vivo evidence, b: Has in-silico evidence.

The second on-target was ERα. It was reported that EA could directly bind to the active pocket

of ERα with high affinity due to its phenolic hydroxyl group and conjugated structure. Pang, et

Page 83: Computational Approach Towards Exploring the

61

al., had measured the binding affinity of ERα to ellagic acid by green Polar Screen ERα

Competitor Assay and found that; ellagic acid had a binding affinity with IC50 value of 62.61 ±

9.34 μM (Pang, et al., 2018).

The third on-target was CDK2. Mohan and Latha had used the SCHRODINGER software to

study the binding features between ellagic acid and CDK2 and had found that EA had high

desirable potential to bind with the active site of CDK2. When CDK2 was docked with EA, the

glide score value was -9.899 Kcl/mol which indicates good interaction with the target protein

(Mohan & Latha, 2018).

CK2 and ERα were discussed in section 4.2.2.1, while in section 4.2.2.4, CDK2 was discussed.

4.2.2.5.2 Off-targets of Ellagic acid

In order to validate the identified potential off-target, in-silico molecular docking simulations

were conducted to reveal the possibility of binding of EA with its new target. EA was

successfully docked within Tank2, PIM1, PDK1 and 17β-HSD1 with a relatively high good

score as shown in table 4.6. This finding could promote further studies on these new off-targets

which could give rise to the discovery of new anticancer/antidiabetic agents against these

important targets.

For mouse GLO1, Tank2 and PIM1, they were discussed in section 4.2.2.1. PDK1 kinase (3-

phosphoinositide-dependent protein kinase-1), is a protein of 556 amino acids belongs to the

family of AGC kinases (protein kinase A (PKA), protein kinase G (PKG), protein kinase C

(PKC)), Although PDK1 was discovered for its ability to phosphorylate protein kinase B (PKB),

also known as AKT, many other kinases are now known to be downstream of PDK1. For

example the AGC kinases serum glucocorticoid dependent kinase (SGK), p70 ribosomal protein

S6 kinases (S6K), p90 ribosomal protein S6 kinase (RSK) and protein kinase C (PKC) isoforms,

are known to be direct targets of PDK1 which phosphorylates specific serine/threonine residues

of their activation loop as seen in figure 4.4. These targets play crucial roles in regulating

physiological processes relevant to metabolism, growth, proliferation and survival For this

reason PDK1 has been named the ―master regulator‖ of AGC kinase signal transduction and

have an important role in the signalling pathways activated by several growth factors and

hormones including insulin signaling (Mora, et al., 2004).

Page 84: Computational Approach Towards Exploring the

65

Increased levels of PDK1 have been reported in 45% of patients with acute myeloid leukaemia,

and a role for PDK1 in breast and ovarian cancer progression has also been proposed. PDK1

inhibitors can be considered as an important therapeutic target in cancer treatment.

Figure ‎11.4: Mechanism of activation of PKB (AKT), S6K and SGK by PDK1. Growth factor or insulin

stimulation activates RTKs which further activates PI3K. Activated PI3K converts PIP2 to PIP3, which provides

docking sites for signaling proteins such as PDK1.Once activated, PDK1 phosphorylates many downstream

effectors, including isoforms of protein kinase B (PKB)/Akt, p70 ribosomal S6 kinase (S6K), serum- and

glucocorticoid-induced protein kinase (SGK) and protein kinase C (PKC), which play crucial roles in regulating

physiological processes relevant to metabolism, growth, proliferation and survival. PTEN functionally antagonizes

PI3K activity by dephosphorylating PIP3 (Raimondi & Falasca, 2011).

On the other hand, PDK1 directly phosphorylates AKT. The activated AKT phosphorylates

many downstream substrates in various signaling pathways, making it a key node in insulin

signaling (Petersen & Shulman, 2018). The activated insulin signaling decreases glucose

production, increases glycogen synthesis, and also increases glucose uptake into peripheral

tissues such as skeletal muscle and adipose tissue as shown in Figure 4.5. The dysfunction of

insulin signaling will cause insulin resistance, which is closely linked to many pathways

Page 85: Computational Approach Towards Exploring the

66

including lipid metabolism, energy expenditure, and inflammation ( Guo, et al., 2020). For this,

PDK1 could be considered as a potential target for insulin resistance and diabetes treatment.

Although extensive data showed that PDK1 plays an important role in cancer, few PDK1-

specific inhibitors have been developed so far. This is largely due to the promiscuity within the

AGC kinase family which makes the design of specific ATP active site-directed inhibitors

difficult (Raimondi & Falasca, 2011).

New specific and potent inhibitors such as GSK2334470 and a pyridinonyl-based compound

were recently characterized and show high selectivity and high potency in inhibiting PDK1.

Interestingly, GSK2334470 inhibits S6K and SGK more potently than Akt. GSK2334470

inhibits PDK1 with an IC50 of ~10 nM, but does not suppress the activity of 93 other protein

kinases. However in the future it will be important to investigate in-vivo the pharmacological

inhibition of PDK1 on tumour models, and determine the pharmacokinetic and metabolic

properties of specific PDK1 inhibitors identified for preclinical trials (Nagashima, et al., 2011).

Figure ‎11.5: Insulin signaling. Insulin binds and activates insulin receptor (INSR), causing phosphorylation of

insulin receptor substrate (IRS). Tyrosine phosphorylated IRS proteins recruit phosphatidylinositide-3 (PI3K),

which catalyzes the production of phosphatidylinositol-3, 4, 5-tris-phosphate (PIP3) from PIP2. PIP3 then activates

PDK1, which phosphorylates activating protein kinase B (AKT). These effector proteins mediate the effects of

insulin on glucose production, utilization, and uptake, as well as glycogen synthesis ( Guo, et al., 2020).

The last off-target for EA was 17β-HSD1, which is a protein composed of 328 amino acids with

a molecular mass of 34.95 kDa. It is primarily expressed in the placenta and ovary, but it is also

Page 86: Computational Approach Towards Exploring the

67

expressed at lower levels in breast epithelium. It is a steroid-converting enzyme that has long

been known to play critical roles in estradiol synthesis and more recently in dihydrotestosterone

inactivation, showing a dual function that promotes breast cancer cell proliferation (Hilborn, et

al., 2017). Recent studies show that 17β-HSD1 increases breast cancer cell migration and

stimulates breast cancer cell growth (Aka, et al., 2012). These findings revealed that, 17β-HSD1

inhibition could be considered as an attractive target of cancer treatment.

4.2.2.6 Ferruginol

Ferruginol is a natural phenol and a meroterpene (a chemical compound containing a terpenoid

substructure) (as seen in table 2.3). It displays relevant pharmacological properties, including

antimicrobial, cardioprotective, antioxidative, antiplasmodial, leishmanicidal, antiulcerogenic,

anti-inflammatory and anticancer (Areche, et al., 2008). It has been shown to inhibit the growth

of cancer cells such as prostate cancer and nonsmall lung cancer (Ho, et al., 2015; Luo,er al.,

2019).

Using RTFA and ferruginol as query template, we had discovered several targets that were

related to cancer, diabetes and hyperlipidemia where all of them were off-targets as in table 4.7.

Table ‎11.7: Pharmacological profiling for ferruginol using ROCS

Target Disease Tanimoto Coefficient Docking Score

HSP90-α Cancer 1.228 -10.621

Tank2 Cancer 1.200 -12.532

CDK2 Cancer 1.138 -8.616

CK2 Cancer 1.118 -9.127

PTP1B T2DM & Hyperlipidemia 1.084 -4.047

PIK3CG Cancer 1.081 -9.560

CDK2: Cycline dependent kinase 2, CK2: Casine kinase 2, HSP90-α: Heat shock protein 90 alpha,

PIK3CG:Phosphatidylinositol-4,5-bisphosphat 3-kinase Catalytic Subunit Gamma, PTP1B: Protein tyrosine

phosphatase1B, Tank2: Tankyrase2.

The first off-target for ferruginol was the human Hsp90 (heat shock protein 90), which is a

chaperone protein that assists other proteins to fold properly, stabilizes proteins against heat

stress, and aids in protein degradation. The mammalian HSP90 family of proteins is a cluster of

highly conserved molecules that are involved in myriad cellular processes. Their distribution in

various cellular compartments underlines their essential roles in cellular homeostasis. HSP90 and

its co-chaperones orchestrate crucial physiological processes such as cell survival, cell cycle

Page 87: Computational Approach Towards Exploring the

68

control, hormone signaling, and apoptosis. Conversely, HSP90, contribute to the development

and progress of serious pathologies, including cancer and neurodegenerative diseases. Therefore,

targeting HSP90 is an attractive strategy for the treatment of neoplasms and other diseases

(Hoter, et al., 2018)

The fifth off-target for ferruginol was PTP1B which is type 1 trans-membrane protein that

catalyze tyrosine phosphorylated proteins, and is widely expressed in different organs in the

human body and involved in different signal transduction pathways including insulin signaling

pathway. It was reported that the binding of α subunits of the insulin receptor lead to a series of

phosphorylation and dephosphorylation cascade reactions including MAPK and PI3K/Akt signal

pathways to regulate metabolism. During the combination of insulin and its receptor, PTP1B

could catalyze insulin receptor and insulin receptor substrates (IRS) de-phosphorylation, which

resulted in down-regulation of insulin signal transduction. Besides, PTP1B could

dephosphorylate activated JAK2 and STAT3, and prevented leptin signal transduction as seen in

Figure 4.6. High expression of PTP1B influenced the activity of PTKs, which resulted in insulin

failing to combine with insulin receptor, induced the IR and leptin resistance, and caused T2DM

and obesity (Zhang & Lee, 2003; Sun, 2016; Abdelsalam et al., 2019).

Figure ‎11.6: The physiological signal pathways involving PTP1B (IR: Insulin receptor, IRS:Insulin receptor

substrate, AKT: Serine/threonine-specific protein kinase, PTP1B: Protein tyrosine phosphatase 1B, JAK2: Janus

kinase 2, STAT3: Signal transducer and activator of transcription 3) (Sun, 2016).

Page 88: Computational Approach Towards Exploring the

69

Genetically modified mice that lack PTP1B protein expression show enhancing in insulin

signaling and glucose tolerance, also they are protected against weight gain and have

significantly lower triglyceride levels when placed on a high-fat diet. This is likely to be

associated with increased energy expenditure owing to enhanced leptin sensitivity (Zhang &

Lee,2003; Sun, 2016).

ISI-113175 (ISIS Pharmaceuticals), an antisense oligonucleotide PTP1B inhibitor (100–200 mg

injected weekly), has completed a phase 2 trial in patients with T2DM on stable, maximal doses

of sulfonyl urea. After 13 weeks, the 200 mg/week cohort reported a 25 mg/dL decrease in

average weekly fasting self-monitoring of blood glucose values (P = 0.026 vs. placebo) and a

significant 65% increase in adiponectin was noted with ISI-113175 (Wang, et al., 2012).

PTP1B is an attractive target for T2DM and hyperlipidemia treatment and this encourage us to

choose it for further investigation. Figure 4.7 shows the binding between feruginol and PTP1B at

molecular level with the presense of hydrogen bonding with key amino acids at the binding site

of PTP1B as ASP181. In addition to the hydrophobic interactions between aromatic rings

composed of TYR46 and PHE182.

Figure ‎11.7: Detailed views of docked ferruginol and the corresponding interacting amino acids within the

binding site of PTP1B (Green lines refere to hydrogen bonds and black lines refere to hydrophobic

interactions).

The last off-target was the human Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic

subunit gamma isoform, PIK3CG (also kown as PI3Kγ) which belonges to 1B PI3K

(Phosphatidylinositol 3-kinases) class. PIK3CG is altered in 2.42% of all cancers with lung

adenocarcinoma, colon adenocarcinoma, cutaneous melanoma, breast invasive ductal carcinoma,

Page 89: Computational Approach Towards Exploring the

71

and conventional glioblastoma multiforme having the greatest prevalence of alterations

(Suvarna, et al., 2017).

PI3K is a group of plasma membrane-associated lipid kinases that exhibits a crucial role in cell

cycle, programmed cell death, DNA repair, angiogenesis, autophagy, motility, and cellular

metabolism (Suvarna, et al., 2017). PI3K consists of three subunits: p85 regulatory subunit, p55

regulatory subunit and p110 catalytic subunit. According to their different structures and specific

substrates, PI3K is divided into 3 classes: classes I, II,and III . Class I PI3Ks comprised of class

IA and class IB PI3Ks. Class IA PI3K, a heterodimer of p58 regulatory subunit and p110

catalytic subunit, is the type most clearly implicated in human cancer. Class IA PI3K contains

p110α, p110β and p110δ catalytic subunits produced from different genes (PIK3CA, PIK3CB

and PIK3CD, respectively), while p110γ produced by PIK3CG represents the only catalytic

subunit in class IB PI3K (Liu, et al., 2009).

PI3Ks have a crucial role in cell cycle, programmed cell death, DNA repair, angiogenesis,

autophagy, motility, and cellular metabolism. It is a potential and druggable target for cancer

therapy. Literature suggests that PI3K signaling pathway is activated in almost 30–50% of

various human cancers. Multiple components of the PI3K signaling pathway are activated and

mutated in human cancers as shown in figure 4.8 (Fruman, et al., 2017; Wang, et al., 2019).

In the last five years, four of the PI3K inhibitors Idelalisib, Copanlisib, Duvelisib and Alpelisib

were approved by the FDA for the treatment of different types of cancer and several other PI3K

inhibitors are currently under clinical development (Bheemanaboina, et al., 2020).

In order to validate the identified potential off-target, in silico molecular docking simulations

were conducted to reveal the possibility of binding of ferruginol with its new targets. The

obtained results revealed that ferruginol was successfully docked within the active site of off-

targets: HSP 90α, Tank2, CDK2, CK2, PTP1B and PIK3CG with relatively good scores as seen

in table 4.7. These findings could promote further studies on these new off-targets which could

give rise to the discovery of new anticancer/antidiabetic agents.

7.7.7.1 Genkwanin

Genkwanin is a monomethoxyflavone that is apigenin in which the hydroxy group at

position 7 is methylated (as shown in table 2.2). Previous pharmacological studies have

found that genkwanin has a variety of pharmacological effects including antibacterial,

Page 90: Computational Approach Towards Exploring the

74

radical scavenging, chemopreventive and inhibiting 17α-Hydroxysteroidsteroid

dehydrogenase type 1 activities (Gao, et al., 2014).

Using RTFA protocol, we had discovered several targets that were related to cancer, diabetes

and hyperlipidemia and all of them were off-targets as shown in table 4.8.

Figure ‎11.8: The overview of PI3K/AKT/mTOR signaling pathway. In physiologic conditions, PI3K is

normally activated by a variety of extracellular stimuli, such as growth factors, cytokines, and hormones,through the

activation of RTK. PIKCG (PI3Kγ) can also be activated by GPCRs. Activated PI3K converts PIP2 to PIP3, which

provides docking sites for signaling proteins such as PDK1 and serine-threonine kinase AKT. Once activated, AKT

phosphorylates many downstream effectors to regulate cell processes such as protein synthesis, cell survival,

proliferation, and metabolism. PTEN functionally antagonizes PI3K activity by dephosphorylating PIP3.

PIP2: Phosphatidylinosito 4,5 phosphatase, PIP3: Phosphatidylinosito 3,4,5 phosphatase, RTK: Receptor tyrosine

kinase, GPCR: G-Protin coupled receptor (Yang, et al., 2019).

All of these off-targets were discussed in pervious sections where PIM1,GP, Tank2, GLO1 and

ERα were illustrated in section 4.2.2.1 while details about PPARγ and HSP90α were presented in

sections 4.2.2.3 and 4.2.2.6 respectively .

For validation of the identified potential off-target, in-silico molecular docking simulations were

used to uncover the possibility of binding of Genkwanin with its new targets. The obtained

Page 91: Computational Approach Towards Exploring the

72

results appeared that the captured off-targets of Genkwanin were successfully docked within the

active site of these proteins with the relatively good score as seen in table 4.8. These findings

could promote further studies on these new off-targets which could give rise to the discovery of

new anticancer/antidiabetic agents.

Table ‎11.8: Pharmacological profiling for Genkwanin using ROCS

Target Disease Tanimoto Coefficient Docking Score

PIM1 Cancer 1.760 -12.754

PPAR-γ T2DM 1.730 -10.489

GP T2DM 1.686 -11.585

Tank2 Cancer 1.576 -15.911

mGLO1 Cancer 1.556 -03.608

ERα Cancer 1.483 -13.907

HSP90-α Cancer 1.330 -12.431

ERα: Estrogen receptor alpha, GP: Glycogen phosphorylase, HSP90-α: Heat shock protein 90 alpha, mGLO1 :

mouse Glyoxalase 1, PIM1: Proviral integration site for moloney murine leukaemia virus 1, PPAR-γ :

Peroxisome proliferator activated receptor gamma, Tank2 : Tankyrase 2.

The docked Genkwanin at the binding site of PIM1 had been illustrated at figure 4.9. The high

similarity (tanimotto coefficient is relatively high) had been supported by the good fitting and

valid bonding between Genkwanin and its predicted target by docking simulations using FRED

software. As shown in the figure, the presence of hydroxyl groups, ketone and methoxy group

produce a potential network of hydrogen bonding with key amino acids at the binding site of

ASP186, SER46 and LYS67, similar to the co-crystallized structure. Moreover, several

hydrophobic interactions are seen between aromatic rings of genkwanin and LEU174 and

ILE185 amino acids.

7.7.7.4 Hispidulin

Hispidulin (4′, 5, 7-trihydroxy-6-methoxyflavone or 6-methoxyapigenin) is a flavones derivative

(as seen in table 2.2) found in several plants. Hispidulin, a bioactive flavone, has been reported

as an effective antioxidant, antifungal, anti-inflammatory and anticancer agent (Patel, et al.,

2016).

By the application of RTFA on Hispidulin as query template, several targets related to cancer

and diabetes were identified, some of them were on-targets such as PIM1 and GP while others

were off-targets as shown in table 4.9.

Page 92: Computational Approach Towards Exploring the

72

A

B

C

Figure ‎11.9: A: Detailed view of docked genkwanin and the corresponding interacting amino acid within the

binding site of PIM1, B: Co-crystallized structure (HUL, PDB code: 4XH6) and the corresponding

interacting amino acids within the binding site of PIM1, C: Overlay view of docked genkwanin within the

binding site of PIM1 (Green lines refere to hydrogen bonds and black lines refere to hydrophobic

interactions).

Page 93: Computational Approach Towards Exploring the

71

7.7.7.4.7 On-targets of Hispidulin

Here we have two on-targets for hispidulin PIM1 and GP and both of them were discussed

previously in section 4.2.2.1.

Crystallographic analysis of PIM1 bound to hispidulin by Chao, et al., 2015 reveals that

hispidulin exhibited PIM1 kinase inhibitory activity with IC50 value of 2.71 μM. According to

the crystal structure of PIM1 kinase in complex with hispidulin, the A ring of hispidulin binds

deep inside the ATP-binding pocket where the methoxy group makes a van der Waals contact

with the hydrophobic residue LEU120. The 7-hydroxyl group and the oxygen of the 6-methoxy

form hydrogen bonds with the highly conserved residues LYS67. Additionally, the oxygen of the

6-methoxy interacts indirectly with PHE187 of the backbone and GLU89 of the side chain

through a water molecule (Chao, et al., 2015).

Table ‎11.9: Pharmacological profiling for hispidulin using ROCS

Target Disease Tanimoto

Coefficient DockingScore Ref.

PIM1 ab

Cancer 2.000 Chao, et al., 2015

mGLOI Cancer 1.730 -2.491

GP b T2DM 1.673 Nath Choudary, 201 4

ERα Cancer 1.427 -14.045

ERβ Cancer 1.360 -13.858

HSP90-α Cancer 1.357 -12.567

ERα: Estrogen receptor alpha, ERβ : Estrogen receptor beta, GP: Glycogen phosphorylase , HSP90-α: Heat

shock protein, mGLO1: mouse Glyoxalase 1, PIM1: Proviral integration site for moloney murine leukaemia

virus 1.

a: Has in-vitro or in-vivo evidence,b: Has in-silico evidence.

The second on-target was GP. In-silico docking study of the hispidulin with GP revealed that

hispidulin had a docking score of -21.189 Kcal/mol. This result showed the high potentiality of

hispidulin to become potential antidiabetic drug ( Nath, Choudhury, 2014).

4.2.2.8.2 Off-targets of Hispidulin

The off-targets that identified by RTFA for hispidulin were CK2α, GLOI, ERα, ERβ and HSP90-

α. All of them discussed before in section 4.2.2.1 except the last target which was discussed in

section 4.2.2.6.

Page 94: Computational Approach Towards Exploring the

75

For validation of the identified potential off-target, molecular docking simulations were used to

uncover the possibility of binding of hispidulin with its new off-targets. The obtained results

releaved that Hispidulin was successfully docked within the active site of these proteins; CK2α,

GLOI, ERα, ERβ and HSP90-α with a relatively good score as seen in table 4.9. These findings

could promote further studies on these new off-targets which could give rise to the discovery of

new anticancer/antidiabetic agents.

4.2.2.9 Luteolin

Luteolin is a tetrahydroxyflavone in which the four hydroxy groups are located at positions 3', 4'

5 and 7 (as shown in table 2.2). It is thought to play an important role in the human body as an

antioxidant, a free radical scavenger, an inflammatory agent and an immune system modulator as

well as being active against several cancers (Chen, et al., 2015).

Using RTFA protocol, luteolin had several targets related to cancer and diabetes, all of them

were on targets: PPAR-γ, Tank2, CK2, PIM1, ERα and HSP90-α exept PDK1which was off-

target as seen in table 4.10.

Table ‎11.10: Pharmacological profiling for luteolin using ROCS

Target Disease Tanimoto

Coefficient Docking score Ref.

PPAR-γ ab

T2DM 2.000 Wang., et al., 2014

Tank2 b Cancer 1.826 Pai, et al., 2017-

CK2 a Cancer 1.826 Lolli, et al.,2012

PIM1a Cancer 1.712

Gadewal & Varma,

2012; Rathod H. 2016.

ERα ab

Cancer 1.429 Maruthanila, et al.,

2019.

HSP90-α b Cancer 1.357 Fu, et al., 2012.

PDK1 Cancer & T2DM 1.297 -12.693

CK2: Casine kinase 2, ERα: Estrogen receptor alpha, HSP90-α: Heat shock protein90 alpha, PDK1:

Phosphoinositide-dependent kinase-1, PIM1: Proviral integration site for moloney murine leukaemia virus 1,

PPAR-γ: Peroxisome proliferator activated receptor gamma, Tank 2: Tankyrase 2.

a: Has in-vitro or in-vivo evidence, b: Has in-silico evidence.

4.2.2.9.1 On-Targets of Luteolin

The first target was PPAR-γ which bind with luteolin with high tanimotto coefficient (2), this

means that luteolin itself was the ligand co-crystallized with PPAR-γ in the PDB. In literature,

Wang, et al., 2014, had documented that luteolin bound to the PPAR-γ with IC50 of 3.9 µM

Page 95: Computational Approach Towards Exploring the

76

forming hydrogen bonds with LYS265 and HIS266 and builds hydrophobic contacts with

various amino acids (Wang, et al., 2014).

The second on-target was Tank2. Luteolin was subjected to the molecular docking process to

identify its binding with Tank2 and the result was promising with a dock score of -11.472

Kcal/mole. The results showed that luteolin had the ability to form additional hydrogen bonding

interactions at the active site of Tank2. The incorporation of hydrogen bond donor or acceptor

groups might increase the binding affinity at the active site of Tank2 (Pai, et al., 2017).

The third target was CK2α. Lolli et al., assayed a panel of 16 flavonoids and related compounds

for their ability to inhibit CK2α. Among the tested compounds, luteolin had an inhibition activity

with IC50 value of 0.50 μM (Lolli, et al., 2012).

The fourth target was PIM1. Gadewal & Varma, investigate the binding of PIM1 and luteolin

and found that luteolin exhibited PIM1 kinase inhibitory activity with IC50 value of 1.6 µM

(Gadewal & Varma, 2012), this support our finding by the RTFA which identified PIM 1 as an

anticancer target.

The fifth target was ERα. In-silico molecular docking study revealed that luteolin showed a high

Glide Score of -8.87 Kcal mol-1

. In addition, Luteolin showed a significant IC50 value of 58.3 ±

4.4 µM against MCF-7 cell line (Maruthanila, et al., 2019).

The last target was HSP90-α. The molecular modeling analysis with CHARMm–Discovery

Studio 2.1 indicated that luteolin could bind to the ATP binding pocket of HSP90. The SPR

technology-based binding assay confirmed the association between luteolin and HSP90. ATP-

sepharose binding assay displayed that luteolin inhibited HSP90-ATP binding (Fu, et al., 2012).

4.2.2.9.2 Off-targets of Luteolin

PDK1 was identified by RTFA as potential off-target and was discussed in section 4.2.2.5.2. For

the validation of this target, docking simulations were conducted to reveal the possibility of

binding of Luteolin with PDK1. Luteolin was successfully docked within the active site of PDK1

with a relatively good score (-12.693). These findings could promote further studies on these

new off-targets which could give rise to the discovery of new anticancer agents.

Page 96: Computational Approach Towards Exploring the

77

4.2.2.10 Oleanolic acid

Oleanolic acid (OA) is an oleanane-type pentacyclic triterpenoid ( as seen in table 2.3) that exist

widely in food, medicinal herbs and other plants. OA exists in nature as a free acid or as an

aglycone of triterpenoids and it is often ubiquitously found with its isomer, ursolic acid. It has

potent pharmacological activities such as antioxidant, anticancer, anti-inflammatory,

antidiabetic, antimicrobial and hepatoprotective effect (Ayeleso, et al., 2017; Ziberna, et al.,

2017).

Using of RTFA protocol on OA as query template, several targets related to cancer and diabetes

were identified, some of them were on-targets as CDK2, PPAR-γ and ERα while others were

off-targets as shown in table 4.11.

4.2.2.10.1 On-Targets of Oleanolic acid

CDK2 was the first on-target. Kim, et al., 2018 evaluated OA role in arresting cell cycle in breast

cancer cells and found the IC50 values for OA induced cytotoxicity was of 132.29 μg/mL. They

found that the treatment of the breast cancer cells with 100 µg/mL OA decreased the production

of CDK2 by 12.02 fold, when compared with that reported for the control group (Kim, et al.,

2018).

Table ‎11.11: Pharmacological profiling for oleanolic using ROCS

Target Disease Tanimoto

Cofficient Docking score Ref.

RPTP-γ Cancer 1.050 -7.871

CDK2 a Cancer 1.011 Kim, et al., 2018

PPAR-γ b T2DM 0.995 Salazar, et al., 2020

ERα b Cancer 0.972 Xie, et al., 2019

17β-HSD1 Cancer 0.971 -12.661

Tank2 Cancer 0.965 -9.238

Mcl-1 Cancer 0.951 -7.384

17β-HSD1: 17 Beta Hydroxysteroid dehydrogenase 1, CDK2: Cycline dependent kinase 2, ERα: Estrogen

receptor alpha, Mcl-1: Myeloid cell leukemia 1, PPAR-γ: Peroxisome proliferatoe activated receptor gamma,

RPTP-γ: Receptor protein tyrosine phosphatase gamma, Tank2: Tankyrase 2.

a: Has in-vitro or in- vivo evidence, b: Has in- silico evidence.

Page 97: Computational Approach Towards Exploring the

78

The second target was PPAR-γ. A study by Salazar, et al., 2020 showed that OA exhibited a

good theoretical affinity against PPAR-α and PPAR-γ with Moldock score values of −118.9 and

−114.2 respectivelly (Salazar, et al., 2020).

Xie, et al., 2019 investigated the binding of OA acid with ERα, their study showed that OA

binds the ERα and up-regulates the expression of ERα on gene and protein levels. In addition,

they docked OA with ERα and found that OA and estradiol have the same interaction site with

ERα, forming intermolecular forces between 3-OH-group of OA and LEU387 of ERα (Xie, et

al., 2019).

4.2.2.10.2 Off-targets of Oleanolic acid

In order to validate the identified potential off-target, docking simulations were conducted to

reveal the possibility of binding of OA with its newly discovered targets; RPTP-γ, 17β-HSD1,

Tank2 and Mcl-1. OA was successfully docked within the active site of these proteins with the

relatively good score as shown in Table 4.11. These findings could promote further studies on

these new off-targets which could be basic nucleus for further optimization that allow the

discovery of new anticancer/ antidiabetic agents.

All of these off-targets of OA were discussed in sections 4.2.2.4, 4.2.2.5.2 and 4.2.2.1

respectively except Mcl-1, which will be discussed here.

Myeloid cell leukemia 1 (Mcl-1) is an anti-apoptotic member of the B-cell lymphoma 2 (Bcl-2)

family of proteins that regulates apoptosis. The Bcl-2 family represented a new class of

oncogenes that promoted oncogenesis, not through upregulation of proliferation, but by

maintaining viability through inhibition of apoptosis. As predicted, dysregulation of Bcl-2

protein family expression and function has since been implicated in virtually all malignancies,

and a number of other pathologies. Mcl-1 blocks the progression of apoptosis by binding and

sequestering the pro-apoptotic proteins Bcl-2 homologous antagonist killer (Bak) and Bcl-2-

associated protein X (Bax), which are capable of forming pores in the mitochondrial membrane,

allowing the release of cytochrome c into the cytoplasm. In the cytoplasm, cytochrome c induces

the activation of a family of cysteine proteases named caspases which are responsible for much

of the macromolecular degradation observed during apoptosis (Thomas, et al., 2010).

Elevated levels of Mcl-1 contribute to tumorigenesis and resistance, not only to conventional

chemotherapies but also to targeted therapies. Accordingly, researchers in both the

Page 98: Computational Approach Towards Exploring the

79

pharmaceutical industry and academia have been actively seeking Mcl-1 inhibitors in the quest

for new anticancer drugs (Fletcher, 2019).

4.2.2.11 Quercetin

Quercetin is a pentahydroxyflavone having the five hydroxy groups placed at the 3-, 3'-, 4'-, 5-

and 7-positions (as shown in table 2.2). It is one of the most abundant flavonoids in edible

vegetables, fruit and wine. It has numerous biological and pharmacological activates as antiviral,

antibacterial, antioxidant, a protein kinase inhibitor, a phytoestrogen, a radical scavenger, a

chelator, an aurora kinase inhibitor, anticancer and many more. In addition, it produces anti-

inflammatory and anti-allergy effects mediated through the inhibition of the lipoxygenase and

cyclooxygenase pathways, thereby preventing the production of pro-inflammatory mediators

(Batiha, et al., 2020).

By using RTFA, we had discovered many targets that were related to cancer and diabetes had

been caught by quercetin, all of them were on-targets as seen in table 4.12. This reflects the

importance of quercetin and the interest in searching its different biological activates.

Table ‎11.12: Pharmacological profiling for quercetin using ROCS

Target Disease Tanimoto

Coefficient Ref.

PPAR-γ b T2DM 1.845 Srinivasan, et al., 2018

CK2 a Cancer 1.692 Lolli, et al.,2012

MAPK14 b Cancer 1.650 Baby, et al., 2016

PIM1 a Cancer 1.597 Bullock, et al., 2005

CDK2 b Cancer 1.340 Baby, et al., 2016

ERα b Cancer 1.306 Maruthanila, et al., 2019

HSP90-α b Cancer 1.277 Kıyga, et al., 2020; Singh, et al., 2015

DAPK1 a Cancer 1.266 Yokoyama, et al., 2015

ERβ b Cancer 1.212 Powers & Setzer, (2015)

CDK 2: Cycline dependent kinase 2, CK2: Casine kinase 2, DAPK1: Death associated protein kinase 1, ERα:

Estrogen receptor alpha, ERβ: Estrogen receptor beta, HSP90-α: Heat shock protein90 alpha, MAPK14:

Mitogen activated protein kinase 14, PIM1: Proviral integration site for moloney murine leukaemia virus 1,

PPAR-γ: Peroxisome proliferator activated receptor gamma.

a Has in-vitro or in-vivo evidence, b Has in- silico evidence.

Page 99: Computational Approach Towards Exploring the

81

From theses targets, PPAR-γ was discussed in section 4.2.2.3, CDK2 was discussed in section

4.2.2.4, HSp90-α was discussed in section 4.2.2.6, the other targets discussed in section 4.2.2.1

and MAPK1 will be discussed here.

The interactions of PPAR-γ with quercetin were extensively discussed. Srinivasan, et al., 2018

had documented that quercetin was an inhibitor for PPAR-γ with docking score of -6.958

Kcal.mol-1

and had interaction with PPAR-γ amino acid residue TYR473 by forming hydrogen

bonding with the OH-group of quercetin (Srinivasan, et al., 2018).

The second target was CK2. Lolli, et al., evaluated the ability of quercetin to inhibit CK2 and

found that quercetin had a potent inhibition activity with IC50 value of 0.55 μM (Lolli, et al.,

2012).

The third target was MAPK14. The effect of quercetin on MAPK14 had been investigated by

Baby, et al., 2016, they had documented that quercetin had hydrophobic bonding with VAL30,

VAL38, ALA51, ILE84, LEU108, ALA111, LEU167, MET179 and TYR182 with a Glide Score

of −6.69 Kcal/mol and binding energy of 44.85 Kcal/mol (Baby et al., 2016).

Similarly, Baby, et al., had investigated the interactions between quercetin and CDK and found

that quercetin had hydrophobic bonding with THR14, LYS33, LEU83, GLN131, ASP145,

ILE10, VAL18, ALA31 and LEU134 with a Glide Score of −9.33 Kcal/mol and binding energy

of −64.14 Kcal/mol (Baby et al., 2016).

MAPK14 is a 41 kDa protein composed of 360 amino acids. MAPK14, also called p38-α,

belongs to the p38 MAPK family which is composed of four members (MAPK14/p38α,

MAPK11/p38β, MAPK12/p38γ and MAPK13/p38δ). The p38 MAPK family consists of highly

conserved proline-directed serine-threonine protein kinases that are activated by various

environmental stresses, growth factors and proinflammatory cytokines. MAP kinases act as an

integration point for multiple biochemical signals, and are involved in a wide variety of cellular

processes such as inflammation, apoptosis, proliferation, differentiation, transcription regulation

and development as seen in figure 4.10 (Santen, et al., 2002 and Nedunuri, et al., 2016).

The role of p38 MAPK in cancer, heart and neurodegenerative diseases was investigated and

found that this pathway was highly attractive for the development of new therapeutics strategies

to treat these pathologies (Almudena & Carmen, 2010).

Page 100: Computational Approach Towards Exploring the

84

Figure ‎11.10: Signaling through MAPK14 cascade and its role in the regulation of cellular functions.

MAPK14 is involved in signaling pathways triggered by a variety of stimuli such as growth factors, oxidative stress,

UV, cytokines and DNA damage. Depending on the stimulus, different receptors and intermediates (adaptors,

GTPases or kinases) are activated leading to the activation of the p38alpha MAPK cascade which is initiated by

activation of MAPKKKs, followed by activation of MAPKKs (MKK3/6/4), which in turn lead to activation of

MAPK14. Once phosphorylated, MAPK14 phosphorylates a number of cytosolic and nuclear substrates, including

transcription factors, which lead to the control of many cellular responses (Almudena & Carmen, 2010).

The fifth target was PIM1. Bullock, et al., identified quercetin as a potent inhibitor of PIM1

kinase, with a Ki value of 25 nM as determined by Isothermal Titration Calorimetry (ITC) and an

IC50 value of 43 nM in enzyme kinetic assays (Bullock, et al., 2005).

Yokoyama, et al., investigated the binding affinity between quercetin and DAPK1 and found that

quercetin exhibited DAPK1 kinase inhibitory activity with IC50 of 8.9 ± 0.90 µM (Yokoyama, et

al., 2015).

Kıyga, et al., 2020 had investigated the interaction betwwen quercetin and HSP90 and found

that the level of HSP90 was almost depleted due to high-dose quercetin (100 µM) treatment

(Kıyga, et al., 2020). Moreover, quercetin had a total interaction energy with HSP90 of -109.87

KJ/mol and a docking score of -71.27 (Singh, et al., 2015).

Page 101: Computational Approach Towards Exploring the

82

The last target was ERβ, its molecular docking energy with quercetin had a value of −106.0

KJ/mol according to Powers & Setzer, (2015) (Powers & Setzer, 2015).

4.2.2.12 Rutin

Rutin (3, 3′, 4′, 5, 7-pentahydroxyflavone-3-rhamnoglucoside) is a flavonol (as shown in table

2.2), abundantly found in many plants. Rutin, also called as rutoside, quercetin-3-rutinoside, and

sophorin. Chemically it is a glycoside comprising of flavonolic aglycone quercetin along with

disaccharide rutinose. It has demonstrated a number of pharmacological activities, including

antioxidant, cytoprotective, vasoprotective, anticarcinogenic, neuroprotective and cardioprotec-

tive activities (Ganeshpurkar, et al., 2017).

Using RTFA and Rutin as query template, we had discovered several targets that were related to

cancer, all of them were off targets except MAPK 14 as seen in table 4.13.

4.2.2.12.1 On-targets of Rutin

The only potential identified on-target was MAPK14, which was discussed in section 4.2.2.11.

Song, et al., 2018 observed that treatment with rutin (30 mg/kg/day for 3 days) reduced p38

MAPK expression compared with the spinal cord injury group (Song, et al., 2018).

Table ‎11.13: Pharmacological profiling for rutin using ROCS

Target Disease Tanimotto

Coefficient

Docking

Score Ref.

DAPK1 Cancer 0.882 -9.752

ERα Cancer 0.790 -8.733

HSP90-α Cancer 0.778 -14.66

CDK2 Cancer 0.777 -8.595

MAPK14 a Cancer 0.766 Song, et al., 2018

PIM1

Cancer

0.756

-12.697

CDK2: Cycline dependent kinase2, DAPK-1: Death associated protein kinase 1, ERα : Estrogen receptor alpha, HSP90-α: Heat shock protein 90 alpha, MAPK14: Mitogen activated receptor 14, PIM1: Proviral integration

site for moloney murine leukaemia virus 1.

a Has in-vitro or in-vivo evidence.

Page 102: Computational Approach Towards Exploring the

82

4.2.2.12.2 Off-targets of Rutin

The off-targets for rutin were DAPK1, ERα, HSP90-α, CDK2 and PIM1. DAPK1, ERα and

PIM1 were discussed in section 4.2.2.1 while HSP90- α and CDK2 were discussed in sections

4.2.2.6 and 4.2.2.4 respectively.

In order to validate these identified potential off-target, in-silico molecular docking simulations

were conducted to reveal the possibility of binding of Rutin with its new targets. Rutin was

successfully docked on DAPK1, ERα, HSP90-α, CDK2 and PIM1 with a relatively high good

score as seen in Table 4.13. This finding could promote further studies on these new off-targets

which could give rise to the discovery of new anticancer agents .

4.2.2.13 Rosmarinic Acid

Rosmarinic acid (RA), an ester of caffeic acid and 3, 4-dihydrophenyllactic acid (as seen in table

2.2), is a naturally occurring phenylpropanoid that is widespread in the plant kingdom. RA has

been widely investigated and has shown many remarkable biological and pharmacological

activities including antioxidant, anti-inflammatory antiviral, antibacterial, antimicrobial, photo-

protective, anticancer, antidepressant, and possible neuroprotective effects (Al-Dhabi, et al.,

2014)

By using RTFA, we had discovered several potential targets for RA that were related to cancer

and diabetes, all of them were off-targets except PPAR-γ as seen in table 4.14.

Table ‎11.14: Pharmacological profiling for rosmarinic acid using ROCS

Target Disease

Tanimotto

Coefficient

Docking

Score Ref.

Mcl-1 cancers. 1.057 -10.702

PPAR-γ ab

T2DM 1.006 Han, et al., 2017

CDK2 Cancer 0.983 -13.420

PDK1 Caner & T2DM 0.958 -14.355

PKACα Cancer 0.957 -15.842

PIK3CG Cancer 0.956 -11.744

CDK2:Cycline dependent kinase2, Mcl-1:Myeloid cell leukemia 1, PDK1: Phosphoinositide-dependent kinase-

1, PIK3CG: Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform, PKACα: Protein

kinase A catalytic subunit alpha, PPAR-γ: Peroxisome proliferator activated receptor gamma.

a Has in-vitro or in- vivo evidence, b Has in-silico evidence.

Page 103: Computational Approach Towards Exploring the

81

4.2.2.13.1 On-targets of Rosmarinic acid

The only identified on target of RA was PPAR-γ which discussed in section 4.2.2.3. Han, et al.,

2017 performed a molecular docking simulation to analyze the interactions between RA and

PPAR-γ. They found that the dock score of PPAR-γ with RA had a value of 8.9589. To validate

the predicted interactions their further studies showed that RA activated PPAR-γ with IC50 value

of 55.78 μM (Han, et al., 2017).

4.2.2.13.2 Off-targets of Rosmarinic acid

The off-targets for RA, Mcl-1, CDK2, PDK1 and PIK3CG were discussed in sections 4.2.2.10.2,

4.2.2.4, 4.2.2.5.2 and 4.2.2.6 respectively.

The protein kinase A catalytic subunit alpha (PKACα) also known as protein kinase cAMP

(3′,5′-cyclic adenosine monophosphate ) activated catalytic subunit alpha or PKACA.

PKACα is a key regulatory enzyme that in humans is encoded by the PRKACA gene. PKA Cα is

a member of the AGC kinase family (protein kinases A, G, and C) of serine/threonine kinases.

PKACα exists as a tetramer comprised of a regulatory (R) subunit dimer and two catalytic (C)

subunits. Upon binding of two molecules of the second messenger cAMP to each R subunit, a

conformational change in the PKA holoenzyme occurs to release the C subunits. These active

kinases phosphorylate downstream targets to propagate cAMP responsive cell signaling events.

PKACα and the other PKA catalytic subunits (PRKACβ and PRKACγ) undergoes many cellular

functions like cell proliferations, cell cycle regulation, and survival of cells through acting on

many substrates (Turnham & Scott, 2016)

Over expression of extracellular PRKACα causes severe tumorgenesis in different organs

(prostate gland, breast, lungs and pancreas) leading to cancer (Swargam, et al., 2010).

In order to validate the identified potential off-target, in-silico molecular docking simulations

were conducted to reveal the possibility of RA binding with its new off-targets. The obtained

results revealed that RA was successfully docked within the active site of Mcl-1, CDK2, PDK1,

PKACα and PIK3CG with relatively good scores as shown in table 4.14. These findings could

promote further studies on these new off targets which could give rise to the discovery of new

anticancer/antidiabetic agents.

Page 104: Computational Approach Towards Exploring the

85

4.2.2.14 Ursolic Acid

Ursolic acid (UA), 3-beta-3-hydroxy-urs-12-ene-28-oic-acid, is a lipophilic pentacyclic triterpen-

oid (as shown in table 2.2). It is widely found naturally in the peels of fruits, as well as in many

herbs and spices. UA has been confirmed to have several biological and pharmacological effects,

such as anti-inflammatory, antitumor, antiplatelet aggregation, antiviral and anti-Mycobacterium

tuberculosis effects (Lee, et al., 2016).

Using RTFA and UA as query template, we had discovered several targets that were related to

cancer and diabetes where all of them were off-targets except ERα as seen in table 4.15.

Table ‎11.15: Pharmacological profiling for ursolic acid using ROCS

Target Disease Tanimotto

coefficient

Docking

Score Ref.

RPTP-γ Cancer 1.052 -6.653

CDK2 Cancer 1.051 -5.922

PPAR-γ T2DM 1.010 -5.910

ERα a cancer. 0.991 Pang, et al., 2018

Tank2 Cancer 0.983 -9.512

CDK2: Cycline dependent kinast2, ERα: Estrogen receptor alpha, PPAR-γ: Peroxisome proliferator-activated

receptor gamma, RPTP-γ: Receptor protein tyrosine phosphatase gamma, Tank2: Tankyrase.

a : Has in-vitro or in-vivo evidence.

4.2.2.14.1 On-targets of Ursolic acid

The only potential identified on-target of UA was ERα (discussed in section 4.2.2.1), Pang, et al.,

had measured the binding affinity of ERα to UA by green Polar Screen ERα Competitor Assay

and found that UA had a binding affinity with IC50 value of 977.38 ± 125.30 μM, (Pang, et al.,

2018).

4.2.2.14.2 Off- targets of Ursolic acid

To validate the UA identified potential off-target, docking simulations were conducted to detect

the possibility of binding of UA with its new off-targets. The obtained results revealed that UA

was successfully docked within the active site of these off-targets as seen in table 4.15.

Page 105: Computational Approach Towards Exploring the

86

4.3 Polypharmacology of Urtica dioica

U. dioica is the most common species of the Urticaceae family commonly known as Stinging

nettle, invasive weed, and one of the most studied medicinal plants worldwide. It can easily be

found on all temperate regions and growing in all seasons. It has been known since ancient times

for their benefits to the human health. Recently there was a rediscovery of the plant as food and

medicine because of the range of biological activities exhibited such as antirheumatic, anti-

hypertension, anti-infective, immuno-modulatory, antihyperlipidemic, antihyperglycaemic, and

allergy relief. Several studies have also reported its analgesic potential and its role as anti-

aggregating factor, as well as describing its favorable effects on treating benign prostate

hyperplasia (Grauso, et al., 2020).

The main components of U. dioica are terpenoids, phenolics, flavonoids and lignans. These

bioactive compounds have demonstrated the numerous pharmacological effects of U. dioica

especially, anticancer, antidiabetic and hypolipidemic effects. In addition, nettle roots were

largely used in the treatment of benign prostatic hyperplasia, whose activity was imputable to

their high content of lignans, which can bind to sex hormone-binding globulin, thus inhibiting

the interaction with the receptor (Xu, et al., 2019).

4.3.1 Reported anticancer, antidiabetic and hypolipidemic targets of U. dioica

Several suggested mechanisms for U dioica anticancer effects have been reported in the

literature. It was reported that U dioica activates apoptosis through the intrinsic pathway by the

increase in the caspase 3 and caspase 9, and a down regulation of anti-apoptotic Bcl2 (B-cell

lymphoma 2) (Mohammadi, et al., 2016). In addition, U dioica induced the anti-metastatic

pathway by decreasing the expression of matrix metalloproteinases 1, 9 and 13, and increased

expression of E-cadherin (Hodroj, et al., 2020). Also, U dioica reduced the activity of 5α-

reductase enzyme which is the key enzyme involved in testosterone metabolism and hormone-

dependent prostate hyperplasia and prostate cancer (Nahata & Dixit, 2012). Moreover, due to the

presence of large quantities of compounds with anti-oxidant and free radical scavenger

properties, U. dioica, is able to reduce the high level of oxidative stress present in cancerous cells

and exert a chemopreventive function (Wang, et al., 2014).

Similarly, several suggested mechanisms for U. dioica antidiabetic activity was reported and

several studies suggest that the U. dioica works as a PPAR-γ agonistic and possess a low

Page 106: Computational Approach Towards Exploring the

87

inhibitory effect against α-amylase and high inhibitory activity against α-glucosidase (Kianbakht,

et al., 2013).

Regarding to the antihyperlipidemic activity of U dioica, the suggested mechanism include the

reduction of HMG-COA reductase activity (Pourahmadi, et al., 2014).

Using RTFA, we had discovered that, U. dioica natural constituents could bind to PPAR-γ as

oleanolic acid and to matrix metalloproteinases7 (MMp7) as secoisolaricirisenol and Caffeoyl-

malic acid, as well as to pancreatic α-amylase as Caffeoylmalic acid.

As shown in table 4.16, eighteen targets related to cancer (3 of them are on target and 15 are off-

targets), six targets related to diabetes (2 of them are on targets and 4 are off targets) and one off-

target related to antihyperlipidemic activities of U. dioica were identified by the adopted RTFA

protocol.

Table ‎11.16: On-targets and off-targets of U.dioica

Anticancer Targets Antidiabetic Targets Hypolipidemic Targets

On-Targets Off Targets On-Targets Off Targets On-Targets Off Targets

1 MMp7 DAPK1 PPAR-γ PTP1B PTP1B

2 Caspase 3 ERα α Amylase DDP-4

3 Caspase 9 ERβ GSK-3β

4 HSP90-α PDK1

5 CDK2

6 PIM1

7 PDK1

8 CK2

9 RPTP-γ

10 Tank2

11 Mcl-1

12 PKACα

13 EGFR

14 FGFR1

15 GSK-3β

CDK2: cycline dependent kinase2, CK2: casine kinase2, DAPK1: death associated protein kinase 1, DPP-4:

dipeptityl peptidase 4, ERα: estrogen receptor alpha, ERβ: estrogen receptor beta, EGFR: epidermal growth

factor receptor, FGFR1: Fibroblast growth factor receptor1, GSK-3β : glycogen synthase kinase-3 beta, HSP90-

α: heat sock protein 90 alpha, Mcl-1:myloid cell leukemia 1, MPP7: matrix metalloproteinases7, PDK1: phosphoinositide-dependent protein kinase-1, PKACα: protein kinase A catalytic subunit alpha, PPAR-γ:

peroxisome proliferator activated receptor gamma, PTP1B: protein tyrosine phosphatase 1B, PIM1: proviral

integration site for moloney murine leukaemia virus 1, RPTP-γ : receptor type protein tyrosine phosphatase

gamma,Tank2: tankyrase2.

Page 107: Computational Approach Towards Exploring the

88

Analysis of results retrieved from RTFA highlights the importance of this approach in clarifying

the polypharmacology of U. dioica especially anticancer, antidiabetic and hypolipidemic

activities as RTFA clarify the ability of U. dioica natural constituents to bind with reported

anticancer targets (on-targets) such as caspase 3, caspase 9 and MMp7, in addition to bind with

antidiabetic targets (on-targets) as PPAR-γ and α-amylase.

4.3.2 Targets identified using U.dioica constituents as queries in RTFA

As mentioned before in section 2.3.2, the main components of U. dioica are terpenoids,

phenolics, flavonoids and lignans. From these components, ellagic acid, oleanolic acid,

quercetin, rutin and ursolic acid are present in U. dioica and S. officinalis and discussed in

sections 4.2.2.5, 4.2.2.10, 4.2.2.11, 4.2.2.12 and 4.2.2.14 respectively.

4.3.2.1 Caffeoylmalic acid

Caffeoylmalic acid(CA), a hydroxycinnamoyl-malate ester (as seen in table 2.6) existing in

various plants, is a vital antioxidant that has an important role in human health. It has shown to

be relevant for the prevention of cardiovascular disease and breast cancer. Furthermore,

caffeoylmalic acid is a distinctive compound of U. dioica (up to 1.6%), which has been widely

used for centuries in the treatment of disease and disorders, such as rheumatism, eczema,

arthritis, gout and anemia (Li, et al., 2018).

Upon applying RTFA on CA as query template, three targets related to cancer and diabetes were

identified and all of them were off-targets as seen in table 4.17.

Table ‎11.17: Pharmacological profiling for caffeoylmalic acid using ROCS

Target Disease Tanimoto Coefficient Docking Score

CDK2 Cancer 1.054 -12.379

HSP90-α Cancer 0.993 -13.404

PTP1B T2DM & Hyperlipidemia 0.990 -8.279

CDK2: Cycline dependent kinase2, HSP90-α :Heat shock protein 90 alpha, PTP1B: Protein tyrosine phosphatase 1B.

To validate the CA identified potential off-target, molecular docking were conducted to detect

the possibility of binding of CA with its off-targets. The obtained results revealed that CA was

successfully docked within the active site of these off-targets with relatively good scores as seen

in table 4.17. These findings could promote further studies on these off-targets which could give

rise to the discovery of new anticancer/antidiabetic/antihyperlipidemic agents.

Page 108: Computational Approach Towards Exploring the

89

4.3.2.2 Chlorogenic acid

Chlorogenic acid (CGA) (as shown in table 2.6) is one of the most available acids among

phenolic acid compounds which can be naturally found in many plants, especially U. dioica (up

to 0.5%). CGA is an important and biologically active dietary polyphenol, playing several

important and therapeutic roles such as antioxidant activity, antibacterial, hepatoprotective,

cardioprotective, anti-inflammatory, antipyretic, neuroprotective, hepatoprotective, antiobesity,

antiviral, anti-microbial, antihypertension, free radicals scavenger and a central nervous system

stimulator. In addition, it has been found that CGA could modulate lipid metabolism and glucose

in both genetically and healthy metabolic related disorders (Naveed, et al., 2018).

Using RTFA protocol, we had discovered four targets of CGA that were related to cancer, and

all of them were off-targets as shown in table 4.18.

Docking was carried out as in-silico validation for the discovered CGA off-targets. The obtained

results revealed that CGA was successfully docked within the active site of these off-targets with

relatively good scores as seen in table 4.18. These findings could promote further studies on

these new off-targets which could give rise to the discovery of new anticancer agents.

Table ‎11.18 : Pharmacological profiling for chlorogenic acid using ROCS

Target Disease Tanimoto Coefficient Docking Score

17β-HSD 1 Cancer 1.088 -13.439

PKACα Cancer 0.972 -15.051

CDK2 Cancer 0.951 -12.38

PIM1 Cancer 0.946 -12.411

17β-HSD1: 17 Beta Hydroxysteroid dehydrogenase 1, CDK2: cycline dependent protein kinase2, PIM1:

Proviral integration site for moloney murine leukaemia virus 1, PKACα: Protein kinase A catalytic subunit

alpha

4.3.2.3 Isolariciresinol

Isolariciresinol is a lignan that is 5, 6, 7, 8-tetrahydronaphthalen-2-ol (as seen in table 2.6). It is

widely distributed in various food plants. Although the biological functions of lignans remain

unclear, some significant pharmacological efects have been revealed, including antitumor and

antioxidative activities (Sampei, et al., 2018).

Page 109: Computational Approach Towards Exploring the

91

As we had applied our adopted approach on isolariciresinol as query template, several target

related to cancer and diabetes were identified; some of them were on-targets as ERα and ERβ

while others were off-targets as seen in table 4.19.

7.2.7.2.7 On- targets of Isolariciresinol

The first on-target identified for isolariciresinol was ERα. Isolariciresinol was found to bind to

the ERα in the position of an agonist with the Glide energy of -73.095 KJ/mol and the Glide

score of -9.78 and had good interaction with the active site residues of MET421 and MET343

with hydrogen bond distance of 3.19Ao and 3.18A

o respectively (Aishwarya, et al., 2019).

Similarly, Isolariciresinol was found to be docked to the ERβ in the position of an antagonist

with the Glide energy of -56.19 KJ/mol and the Glide score of -8.75 and had good interaction

with the active site residues of ASP258 and LEU353 with hydrogen bond distance of 3.13A° and

2.41A° respectively (Aishwarya, et al., 2019).

Table ‎11.19: Pharmacological profiling for isolariciresinol using ROCS

Target Disease Tanimotto

Coefficient

Docking

Score Ref.

CDK2 Cancer 1.127 -10.755

EGFR Cancer 1.126 -10.586

ERα b Cancer 1.105

Aishwarya, et al.,

2019

17β-HSD 1 Cancer 1.089 -12.596

HSP90-α Cancer 1.05 -13.00

PDK1 Cancer & T2DM 1.006 -11.592

ERβ b Cancer 1.004

Aishwarya, et al.,

2019.

FGFR1 Cancer 1.00 -11.997

17β-HSD 1: 17 Beta Hydroxysteroid dehydrogenase 1, CDK2: Cycline dependent kinase 2, EGFR: Epidermal

growth factor receptor, ERα: estrogen receptor alpha, ERβ: Estrogen receptor beta, FGFR1: Fibroblast growth

factor receptor 1, HSP90-α: Heat shock protein 90 alpha, PDK1: Phosphoinositide dependent protein kinase 1.

b: Has in-silico evidence.

4.3.2.3.2 Off-targets of Isolariciresinol

The identified potential off-targets of Isolariciresinol; CDK2, 17β-HSD1, HSP90-α, PDK1and

FGFR were mentioned previously in sections 4.2.24, 4.2.2.5.2, 4.2.2.6, 4.2.2.5.2 and 4.2.2.2

respectively.

Page 110: Computational Approach Towards Exploring the

94

The EGFR, also referred to human epidermal growth factor receptor HER1/ErbB1, belongs to a

larger family of ErbB receptors with tyrosine kinase activity. The gene symbol, ErbB, is derived

from the name of a viral oncogene to which these receptors are homologous: erythroblastic

leukemia viral oncogene. Other members of the HER family include ErbB2/HER2, ErbB3/HER3

and ErbB4/HER4. Insufficient ErbB signaling in humans is associated with the development of

neurodegenerative diseases, such as multiple sclerosis and Alzheimer's disease (Bublil & Yarden,

2007).

EGFR is frequently overexpressed and/or hyperactivated in human malignancies such as non-

small-cell lung cancer, pancreatic cancer, breast cancer, and colon cancer and therefore EGFR

inhibitors may be used in the treatment of cancers. EGFR overexpression and activation are

known to significantly impact cancer cell hallmark traits, such as increased cell survival,

proliferation and invasion as seen in figure 4.11 (Xu, et al., 2017).

Figure ‎11.11: Epidermal growth factor receptor (EGFR) and its downstream signaling proteins. EGF,

epidermal growth factor; EGFR, epidermal growth factor receptor; JAK, Janus kinase; STAT, signal transducer and

activator of transcription; PI3K, phosphatidylinositol 3-kinase; MAPK, mitogen-activated protein kinase; Akt,

protein kinase B; P, phosphorylation. T3; tocotrienol Arrows and perpendicular lines indicate activation/induction

and inhibition/suppression, respectively (Eitsuka,et al., 2016).

Page 111: Computational Approach Towards Exploring the

92

EGFR inhibitors can be classified as either tyrosine kinase inhibitors (TKI) (eg, erlotinib,

gefitinib): these bind to the tyrosine kinase domain in the epidermal growth factor receptor and

stop the activity of the EGFR or monoclonal antibodies (eg, cetuximab, necitumumab): these

bind to the extracellular component of the EGFR and prevent epidermal growth factor from

binding to its own receptor, therefore preventing cell division (Gerber, 2008; Liang, et al., 2014).

In-silico docking simulations were applied to detect the possibility of binding of Isolariciresinol

with its new targets. The obtained results revealed that Isolariciresinol was successfully docked

within the active site of CDK2, EGFR, 17β-HSD 1, HSP90-α, PDK1 and FGFR1 with relatively

good scores as shown in table 4.19.

The EGFR1 had a good docking score and this encouraged us to study the interaction between

Isolariciresinol and EGFR1 at molecular level as shown in figure 4.12. The figure shows a strong

network of hydrogen bonds between hydroxyl and methoxy groups of Isolariciresinol and key

amino acids in the binding site of EGFR1 including ASP855, LEU788, LEU792, GLN791 and

GLY796 similar to the co-crystallized structure. Moreover, the other aromatic rings occupy a

lipophilic cavity composed of THR854, LUE844 and VAL726.

7.2.7.7 Neoolivil

Neoolivil is the main lignan in the roots of U. dioica. Its chemical structure was shown in table

2.6. It belongs to the class of organic compounds known as 7,7'-epoxylignans. These are lignans

with a structure based on a 2,5-diaryl-3,4-dimethyltetrahydrofuran skeleton. Neoolivil have no

activity reported in the literature, but the biological activites of lignans were well established and

include antioxidant, antitumor, estrogenic, antimicrobial, and cholesterol lowering activities

(Tsopmo, et al., 2013).

Using RTFA ptotocol, we had discovered several targets of neoolivil that were related to cancer

and diabetes and all of them were off-targets as shown in table 4.20.

In-silico target fishing had led to identification of eight new off-targets for neoolivil:ERα, 17B-

HSD1, DPP-4, CDK2, Tank2, ERβ, HSP90-α and GSK-3β. All of these targets have been

discussed before except GSK-3β.

GSK3 is a multifunctional proline-directed serine/threonine kinase. It was named for its ability to

phosphorylate, and thereby inactivate glycogen synthase, a key regulatory molecule in the

synthesis of glycogen. There are two highly homologous forms of GSK3 in mammals encoded

by distinct genes, GSK3α (51 kDa) and GSK3β (47 kDa). Although GSK-3 originally was

identified to have functions in regulation of glycogen synthase, it was subsequently determined

Page 112: Computational Approach Towards Exploring the

92

to have roles in multiple normal biochemical processes as well as various disease conditions as

Alzheimer‘s disease (AD) and mood disorders, osteoporosis, atherosclerosis, cancer and cardiac

hypertrophy (Phukan, 2010).

A

B

C

Figure ‎11.12: A; Detailed view of docked Isolariciresinol and the corresponding interacting amino acid within

the binding site of EGFR, B; Detailed view of co-crystallized structure (1C9 , PDB code: 4I23 ) and the

corresponding interacting amino acid within the binding site of EGFR, C; Overlay view of docked

Isolariciresinol within the binding site of EGFR (Green lines refere to hydrogen bonds and black lines refere

to hydrophobic interactions).

GSK-3 is sometimes referred to as a moonlighting protein due to the multiple substrates and

processes which it controls. Frequently, when GSK-3 phosphorylates proteins, they are targeted

for degradation. GSK-3 is often considered as a component of the PI3K/PTEN/AKT/GSK-

3/mTORC1 pathway as GSK-3 is frequently phosphorylated by AKT which regulates its

Page 113: Computational Approach Towards Exploring the

91

inactivation. AKT is often active in human cancer and hence, GSK-3 is often inactivated (Duda,

et al., 2020).

Recent studies in colorectal cancer, pancreatic cancer, hepatocellular carcinoma and ovarian

cancer demonstrate that GSK-3β is involved in the process of tumorigenesis. Inhibition of the

expression and activity of GSK-3β attenuates cell proliferation and causes apoptosis in

colorectal, pancreatic and ovarian cancer cells (Wang, et al., 2008).

Table ‎11.20: Pharmacological profiling for neoolivil using ROCS

Target Disease Tanimoto Coefficient Docking Score

ERα Cancer 1.18 -12.452

17B-HSD 1 Cancer 1.122 -14.14

DPP-4 T2DM 1.08 -9.07

CDK2 Cancer 1.002 -9.502

Tank2 Cancer 1.00 -12.393

ERβ Cancer 0.997 -9.894

HSP90-α Cancer 0.985 -13.29

GSK-3β T2DM & Cancer 0.978 -10.639

17B-HSD1:17 Beta Hydroxysteroid dehydrogenase 1, DPP-4: Dipeptidyl peptidase 4, ERα :Estrogen receptor

alpha, ERβ: Estrogen receptor beta, GSK-3β : Glycogen synthase kinase-3 beta , HSP90-α: Heat shock protein

90 alpha, Tank2: Tankyrase 2.

Numerous GSK-3 inhibitors were developed by multiple pharmaceutical companies since the

initial characterization of the biochemical effects of GSK-3 and the association of GSK-3 with

many common immunological disorders and cancer (Duda, et al., 2020). For example, Lithium is

a well-known GSK-3 inhibitor was initially and still is used for the treatment of various

neurological disorders including bipolar disorder (manic depression). It can inhibit proliferation

of the human esophageal cancer cell lineas well as other cell lines of different tissue types by

inducing the phosphorylation of GSK-3β which can lead to inactivation of GSK-3 (Wang, et al.,

2008).

The validity of these proteins as potential targets for neoolivil was evaluated by molecular

docking simulation according to our adopted experimental procedures and the obtained results

were shown in table 4.20 and they reflected successful docking for neoolivil with all new off-

targets with relatively good scores.

DPP-4 as a potential target for neoolivil had a Tanimoto coefficient higher than 1 and a good

docking score, this had invited us to ensure the presence of possible interactions between

Page 114: Computational Approach Towards Exploring the

95

neoolivil and DPP-4 at molecular level as shown in figure 4.13. The figure shows the hydrogen

bonding between neoolivil and key amino acids at the binding site of DPP4 as GLU206,

TYR547, TYR662 and TYR666. In addition, the other aromatic rings occupy a lipophilic cavity

composed of TYP659 and PHE357.

A B

Figure ‎11.13: A: Detailed view of docked Neoolovil and the corresponding interacting amino acid within the

binding site of DPP4, B: Surface view of docked Neoolovil and the corresponding interacting amino acid

within the binding site of DPP-4 (Green lines refere to hydrogen bonds and black lines refere to hydrophobic

interactions).

7.2.7.4 Secoisolariciresinol

Secoisolariciresinol is a lignan, a type of phenylpropanoid (seen in table 2.6). Lignans are a

group of phytonutrients which are widely distributed in the plant kingdom. In the intestine the

gut microflora can form secoisolariciresinol from the secoisolariciresinol diglucoside. The

majority of studies demonstrate that secoisolariciresinol diglucoside has various biological

properties including anti-inflammatory, antioxidant, antimutagenic, antimicrobial, antiobesity,

antihypolipidemic and neuroprotective effects (Imran, et al., 2015).

After the application of RTFA protocol on Secoisolariciresinol as query template, we had

discovered four targets that were related to cancer and diabetes and all of them were off-targets

except ERα as shown in table 4.21.

4.3.2.5.1 On-targets of Secoisolariciresinol

ERα was the only off-target identified for secoisolariciresinol. Secoisolariciresinol was found to

Page 115: Computational Approach Towards Exploring the

96

bind to the ERα in the position of an agonist with the Glide energy of -35.144 KJ/mol and Glide

Score of -8.19 (Aishwarya, et al., 2019).

Table ‎11.21: Pharmacological profiling for secoisolariciresinol using ROCS

Target Disease Tanimoto

Coefficient

Docking Score Ref.

HSP90-α Cancer 0.924 -13.025

PKACα Cancer 0.908 -15.728

Tank2 Cancer 0.884 -13.192

ERα b Cancer 0.879

Aishwarya , et al.,

2019.

ERα: Estrogen receptor alpha, HSP90-α : Heat shock protein 90 alpha, PKACα: Protein kinase A catalytic

subunit alpha , Tank 2: Tankyrase 2.

b: Has in-silico evidence.

4.3.2.5.2 Off-targets of Secoisolariciresinol

For the validation of these identified potential off-target, molecular docking were applied to

reveal the possibility of binding of Secoisolariciresinol with its new off-targets. The obtained

results showed that Secoisolariciresinol was successfully docked within the active site of HSP90-

α, PKACα and Tank2 with the relatively good scores as shown in table 4.21. These findings

could promote further studies on these new off-targets which could give rise to the discovery of

new anticancer agents.

12

13

14

15

16

17

18

19

20

21

22

23

Page 116: Computational Approach Towards Exploring the

97

24

25

26 Chapter Five

Conclusion

Salvia officinalis and Urtica dioica exhibits a wide range of biological activities especially

anticancer, antidiabetic and hypolipidemic activitis as the results of our research had been

shown, these activities were attained by modulation of different biological targets.

Targets identification for S. officinalis and U. dioica is a very important step not only for full

understanding of their bioactivity and mechanisms of action, but also for the discovery and

development of new, potent and less toxic drugs for the treatment of serious diseases as cancer,

diabetes and hyperlipidemia.

Herein, the adopted ligand based target fishing approach, RTFA, facilitated the target profiling

of S. officinalis and U. dioica phytochemicals and was able to retrieve well known targets and

several new off-targets that are not reported previously as direct targets and this increases

confidence in our approach in capturing the right positive targets and in exploring the new off-

targets.

The phytochemicals of S. officinalis and U. dioica were successfully docked within active sites

of the captured off-targets as an in-silico evidence

By the application of RTFA on the phytochemicals, the concept of polypharmacology is

obviously clarified as multiple phytochemicals could affect the same target on the disease

pathway producing synergistic effect or an individual compound affecting multiple targets that

involved on the same disease.

27

28

Page 117: Computational Approach Towards Exploring the

98

29

30 Chapter 6

Recommendations

Use of natural products are safe and effective alternatives compared to unsafe drugs.

In-silico studies are rapid, time preserving and relatively accurate methods for drug discovery.

Finally, further studies on the phytochemicals of S. officinalis and U. dioica against the fished

off-targets will provide more understanding of their pharmacological properties as well as could

provide good leads for designing new more potent and safer anticancer, dantidiabetic and

hypolipidemic drugs.

Page 118: Computational Approach Towards Exploring the

99

31 References

Abbas, Z. & Rehman, S. (2018). Neoplasm, Edittor : Hafiz Naveed Shahzad, H. N., Chapter 6 ;

An Overview of Cancer Treatment Modalities, 139- 157.

Abdelsalam, S. S., Korashy, H. M., Zeidan, A., & Agouni, A. (2019). The Role of Protein

Tyrosine Phosphatase (PTP)-1B in Cardiovascular Disease and Its Interplay with Insulin

Resistance. Biomolecules, 9(7), 286.

Abdulhussein, Lina., Allaithi, A., Matrood, W., Al-Azawi, W. (2019). Study Of The Effect Of

Salvia Officinalis Leaves Extract And Xenical Drug On Some Of The Biochemical And

Histological Parameters In The Rats Induced With Hyperlipidemia. Plant Archives, 19(2),

1111-1122.

Achenbach, J., Tiikkainen, P., Franke, L., Proschak, E. (2011). Computational tools for

polypharmacology and repurposing. Future medicinal chemistry, 3(8), 961–968.

Agius, L. (2007). New hepatic targets for glycaemic control in diabetes. Best practice &

research. Clinical endocrinology & metabolism, 21(4), 587–605.

Ahangarpour, A., Mohammadian, M. & Dianat, M. (2012). Antidiabetic effect of hydroalcholic

urticadioica leaf extract in male rats with fructose-induced insulin resistance. Iranian

journal of medical sciences, 37(3), 181–186.

Aishwarya, S., Nargis, B. & Anita, M,. (2019). An in silico study of lignans as selective estrogen

receptor modulators to treat viral infections. International Research. Journal of Biological

Sciences, 8(3), 19-25.

Aka, J.A., Zerradi, M., Houle, (2012). 17beta-hydroxysteroid dehydrogenase type 1 modulates

breast cancer protein profile and impacts cell migration. Breast Cancer Research, 14(3),

92-106.

Akasaka, H., Akasaka, T., Kurata, M., Ueda, C., Shimizu, A., Uchiyama, T., Ohno, H. (2000).

Molecular anatomy of BCL6 translocations revealed by long-distance polymerase chain

reaction-based assays. Cancer research, 60(9), 2335–2341.

Akbay, P., Basaran, A.A., Undeger, U., Basaran, N. (2003). In vitro immunomodulatory activity

of flavonoid glycosides from Urtica dioica L. Phytotherapy Research,17(1), 34-37.

Alchab, F., Ettouati, L., Bouaziz, Z. (2015).Synthesis, Biological Evaluation and Molecular

Modeling of Substituted Indeno[1,2-b]indoles as Inhibitors of Human Protein Kinase CK2.

Pharmaceuticals (Basel, Switzerland),8(2), 279-302.

Page 119: Computational Approach Towards Exploring the

411

Al-Dhabi, N. A., Arasu, M. V., Park, C. H., Park, S. U. (2014). Recent studies on rosmarinic acid

and its biological and pharmacological activities. EXCLI Journal, 13, 1192–1195.

Ali, S., Nasreen, S., Safeer, S., Andleeb, S., Ejaz, M., Bano, S., Shakir, H. (2016). Medicinal

plants as therapeutic agents for cancer treatment. Punjab University Journal of Zoology.

31, 295-305.

Almasri, I.M. (2018). Exploring polypharmacology of some natural products using similarity

search target fishing approach. International Journal of Computational Biology and Drug

Design, 11(4), 295-309.

Al-Menhali, A., Al-Rumaihi, A., Al-Mohammed, H., Al-Mazrooey, H., Al-Shamlan, M.,

AlJassim, M., Al-Korbi, N., Eid, A. H. (2015). Thymus vulgaris (thyme) inhibits

proliferation, adhesion, migration, and invasion of human colorectal cancer cells. Journal

of medicinal food, 18(1), 54–59.

Almudena, P. & Carmen, G. (2010). Atlas of Genetics and Cytogenetics in Oncology and

Haematology, MAPK14 (mitogen-activated protein kinase 14,15(8),628-631.

Alsemari, A., Alkhodairy, F., Aldakan, A., Al-Mohanna, M., Bahoush, E., Shinwari, Z., Alaiya,

A. (2014). The selective cytotoxic anti-cancer properties and proteomic analysis of

Trigonella Foenum-Graecum. Complementary and alternative medicine, 14, 114.

Al-Wasfi, R.M., Jabaar, H.J., Dergham, M., Hameed, A.F.(2012). Studying the hypoglycemic

and the antibacterial activity of various plant extract of Urtica dioica. Al-kufa Journal of

Biology, 4, 232-242.

Amit, G., Vandana, S., Sidharth, M. (2011). Hyperlipidemia: An Updated Review. International

Journal of Biopharmaceutical & Toxicological Research,1(1), 81-89.

Areche, C., Theoduloz, C., Yáñez, T., Souza-Brito, A. R., Barbastefano, V., Paula, D., Ferreira,

A. L., Schmeda-Hirschmann, G., Rodríguez, J. A. (2008). Gastroprotective activity of

ferruginol in mice and rats: effects on gastric secretion, endogenous prostaglandins and

non-protein sulfhydryls. The Journal of pharmacy and pharmacology, 60(2), 245–251.

Arqués, O., Puig, I., Argilés, G., Dienstmann, R., Fernández, N., Caratù, G., Matito, J.,

Silberschmidt, D. (2016). Tankyrase inhibition blocks Wnt/β-catenin pathway and reverts

resistance to PI3K and AKT inhibitors in the treatment of colorectal cancer. Clinical

Cancer Research, 22,644-656.

Arun, K., Sunil, C., Dhaliya, S.A., Surya, A. S., Dawn, V. T., Betty, C. (2013). A Review Of

Hyperlipidemia And Medicinal Plants. International Journal of Applied Pharmaceutical

Sience, 2(4) ,219-237.

Asadi-Samani, M., Kooti, W., Aslani, E., & Shirzad, H. (2016). A Systematic Review of Iran's

Medicinal Plants With Anticancer Effects. Journal of evidence-based complementary &

alternative medicine, 21(2), 143–153.

Page 120: Computational Approach Towards Exploring the

414

Ashburn, T. T. & Thor, K. B. (2004). Drug repositioning: identifying and developing new uses

for existing drugs. Nature reviews. Drug discovery, 3(8), 673–683 .

Ashcroft, F.M. & Rorsman, P. (2012). Diabetes mellitus and the β cell: The last ten years. Cell,

148, 1160–1171.

Atlee, Clement. (2020). Overview of ideal antilipidemic drugs: Past, present and the future. GSC

Biological and Pharmaceutical Sciences. 11(3), 067-074.

Avci, G., Kupeli, E., Eryavuz, A., Yesilada, E., & Kucukkurt, I. (2006).

Antihypercholesterolaemic and antioxidant activity assessment of some plants used as

remedy in Turkish folk medicine. Journal of ethnopharmacology, 107(3), 418–423.

Ayeleso, T. B., Matumba, M. G., Mukwevho, E. (2017). Oleanolic Acid and Its Derivatives:

Biological Activities and Therapeutic Potential in Chronic Diseases. Molecules (Basel,

Switzerland), 22(11), 1915.

Azevedo, M. I., Pereira, A. F., Nogueira, R. B., Rolim, F. E., Brito, G. A., Wong, D. V., Lima-

Júnior, R. C., de Albuquerque Ribeiro, R., & Vale, M. L. (2013). The antioxidant effects of

the flavonoids rutin and quercetin inhibit oxaliplatin-induced chronic painful peripheral

neuropathy. Molecular pain, 9(1), 53-67.

Baba, K., Hiramatsu, R., Suradej, B., Tanigaki, R., Koeda, S., Waku, T., & Kataoka, T. (2018).

Asiatic Acid, Corosolic Acid, and Maslinic Acid Interfere with Intracellular Trafficking

and N-Linked Glycosylation of Intercellular Adhesion Molecule-1. Biological and

Pharmaceutical Bulletin, 41(12), 1757–1768.

Baby, B., Antony, P., Al Halabi, W., Al Homedi, Z., Vijayan, R. (2016). Structural insights into

the polypharmacological activity of quercetin on serine/threonine kinases. Drug design,

development and therapy, 10, 3109–3123.

Badiee P, Nasirzadeh A R and Motaffaf M: (2012) Comparison of Salvia officinalis L. essential

oil and antifungal agents against candida species. Journal of Pharmaceutical Technology

and Drug Research, 1(1),7.

Bahmani, M., Mirhoseini, M., Shirzad, H., Sedighi, M., Shahinfard, N., Rafieian-Kopaei, M.

(2015). A Review on Promising Natural Agents Effective on Hyperlipidemia. Journal of

Evidence-Based Complementary & Alternative Medicine, 20(3), 228–238.

Bai, T., Tanaka, T., Yukawa, K. (2010). Targeted knockdown of death-associated protein kinase

expression induces TRAIL-mediated apoptosis in human endometrial adenocarcinoma

cells. International Journal of Oncology, 37, 203-210.

Baier, A., Nazaruk, J., Galicka, A., Szyszka, R. (2018). Inhibitory influence of natural flavonoids

on human protein kinase CK2 isoforms: effect of the regulatory subunit. Molecular and

Cellular Biochemistry, 444, 35–42 .

Page 121: Computational Approach Towards Exploring the

412

Bailey, C.J. & Day, C. (2004). Metformin: its botanical background. Practical Diabetes

International. 21(3),115–117.

Baker, N. C., Ekins, S., Williams, A. J., Tropsha, A. (2018). A bibliometric review of drug

repurposing. Drug discovery today, 23(3), 661–672.

Balzarini, J., Van Laethem, K., Hatse, S., Froeyen, M., Peumans, W., Van Damme, E., Schols,

D. (2005). Carbohydrate-binding agents cause deletions of highly conserved glycosylation

sites in HIV GP120: a new therapeutic concept to hit the achilles heel of HIV. The Journal

of biological chemistry, 280(49), 41005–41014.

Bao, R., Christova, T., Song, S., Angers, S., Yan, X., & Attisano, L. (2012). Inhibition of

tankyrases induces Axin stabilization and blocks Wnt signalling in breast cancer cells.

PloS one, 7(11), 48670.

Batiha, G. E., Beshbishy, A. M., Ikram, M., Mulla, Z. S., El-Hack, M., Taha, A. E., Algammal,

A. M.,Elewa, Y. (2020). The Pharmacological Activity, Biochemical Properties, and

Pharmacokinetics of the Major Natural Polyphenolic Flavonoid: Quercetin. Foods (Basel,

Switzerland), 9(3), 374 .

BC Cancer Agency. Pharmacy Education Program. Cancer Drug Pharmacology Table 2019-10-

04. (2019). Author: Rhonda Kalyn, BC Cancer Agency. Reviewer: Mario de Lemos, BC

Cancer Agency.

Bheemanaboina, R. (2020). Isoform-Selective PI3K Inhibitors for Various Diseases. Current

topics in medicinal chemistry, 20(12), 1074–1092.

Bnouham, M., Merhfour, F. Z., Ziyyat, A., Mekhfi, H., Aziz, M., Legssyer, A. (2003).

Antihyperglycemic activity of the aqueous extract of Urtica dioica. Fitoterapia, 74(7-8),

677–681.

Bublil, E. M. & Yarden, Y. (2007). The EGF receptor family: spearheading a merger of

signaling and therapeutics. Current opinion in cell biology, 19(2), 124–134.

Bullock, A. N., Debreczeni, J. E., Fedorov, O. Y., Nelson, A., Marsden, B. D., Knapp, S. (2005).

Structural basis of inhibitor specificity of the human protooncogene proviral insertion site

in moloney murine leukemia virus (PIM-1) kinase. Journal of medicinal chemistry, 48(24),

7604–7614.

Burger, M., Lindvall, M., Han, W. (2008). Pim kinase inhibitors and methods of their use.

Neoplasia, 16(5), 403–412.

Burrell, R. A., McGranahan, N., Bartek, J., Swanton, C. (2013). The causes and consequences of

genetic heterogeneity in cancer evolution. Nature, 501(7467), 338–345.

Page 122: Computational Approach Towards Exploring the

412

Busch, A. M., Johnson, K. C., Stan, R. V., Sanglikar, A., Ahmed, Y., Dmitrovsky, E., &

Freemantle, S. J. (2013). Evidence for tankyrases as antineoplastic targets in lung cancer.

BMC cancer, 13, 211.

Carter, E. P., Fearon, A. E., & Grose, R. P. (2015). Careless talk costs lives: fibroblast growth

factor receptor signalling and the consequences of pathway malfunction. Trends in cell

biology, 25(4), 221–233.

Cereto-Massagué, A., Ojeda, M. J., Valls, C., Mulero, M., Pujadas, G., Garcia-Vallve, S. (2015).

Tools for in silico target fishing. Methods (San Diego, Calif.), 71, 98–103.

Chao, S. W., Su, M. Y., Chiou, L. C., Chen, L. C., Chang, C. I., Huang, W. J. (2015). Total

Synthesis of Hispidulin and the Structural Basis for Its Inhibition of Proto-oncogene

Kinase Pim-1. Journal of natural products, 78(8), 1969–1976.

Chen B., Mcconnell, K.J., Wale, N., Wild, D.J., Gifford, E.M. (2011). Comparing bioassay

response and similarity ensemble approaches to probing protein pharmacology.

Bioinformatics, 27, 3044–3049.

Cheshomi, H., Bahrami, A. R., & Matin, M. M. (2020). Ellagic acid and human cancers: a

systems pharmacology and docking study to identify principal hub genes and main

mechanisms of action. Molecular diversity, 10.1007/s11030-020-10101-6.

Christensen, K. B., Jørgensen, M., Kotowska, D., Petersen, R. K., Kristiansen, K., Christensen,

L. P. (2010). Activation of the nuclear receptor PPARγ by metabolites isolated from sage

(Salvia officinalis L.). Journal of ethnopharmacology, 132(1), 127–133 .

Coffman, K., Brodney, M., Cook, J., Lanyon, L., Pandit, J., Sakya, S., Schachter, J., Tseng-

Lovering, E., Wessel, M. (2011). 6-amino-4-(pyrimidin-4-yl)pyridones: novel glycogen

synthase kinase-3β inhibitors. Bioorganic & medicinal chemistry letters, 21(5), 1429–

1433.

Costa, J. (2020, October 15). Cancer. Encyclopedia Britannica.

https://www.britannica.com/science/cancer-disease

Cozza, G., Bonvini, P., Zorzi, E., Poletto, G., Pagano, M. A., Sarno, S., Moro, S. (2006).

Identification of Ellagic Acid as Potent Inhibitor of Protein Kinase CK2: A Successful

Example of a Virtual Screening Application. Journal of Medicinal Chemistry, 49(8), 2363–

2366.

Daher, C. F., Baroody, K. G., & Baroody, G. M. (2006). Effect of Urtica dioica extract intake

upon blood lipid profile in the rats. Fitoterapia, 77(3), 183–188 .

Dakshanamurthy, S., Naiem. T. I., Shahin A., Ashwini, S., Oakland, J. P., Subha, M., Aykut, U.,

Milton, L. B., Stephen W. B. (2012). Predicting new indications for approved drugs using

a proteochemometric method, Jornal of Medicinal Chemistry, 55, 6832–6848.

Page 123: Computational Approach Towards Exploring the

411

Dar, K. B., Bhat, A. H., Amin, S., Hamid, R., Anees, S., Anjum, S., Reshi, B. A., Zargar, M. A.,

Masood, A., Ganie, S. A. (2018). Modern Computational Strategies for Designing Drugs to

Curb Human Diseases. Current topics in medicinal chemistry, 18(31), 2702–2719.

Deacon C. F. (2019). Physiology and Pharmacology of DPP-4 in Glucose Homeostasis and the

Treatment of Type 2 Diabetes. Frontiers in endocrinology, 10, 80.

DeFronzo, R. A., Triplitt, C. L., Abdul-Ghani, M., Cersosimo, E. (2014). Novel Agents for the

Treatment of Type 2 Diabetes. Diabetes spectrum : A Publication of the American

Diabetes Association, 27(2), 100–112.

Dizaye, K., Alberzingi, B.O., & Sulaiman, S. (2013). Renal and vascular studies of aqueous

extract of Urtica dioica in rats and rabbits. Iraqi Journal of Veterinary Sciences, 27, 25-31.

Duda, P., Akula, S. M., Abrams, S. L., Steelman, L. S., Martelli, A. M., Cocco, L., Ratti, S.,

Candido, S., Libra, M., Montalto, G., Cervello, M., Gizak, A., Rakus, D., McCubrey, J. A.

(2020). Targeting GSK3 and Associated Signaling Pathways Involved in Cancer. Cells,

9(5), 1110.

Early Breast Cancer Trialists‘ Collaborative Group (EBCTCG). (2005).Effects of chemotherapy

and hormonal therapy for early breast cancer on recurrence and 15-year survival: An

overview of the randomised trials. The Lancet. 365(9472),1687-1717.

Edmondson, S. D., Mastracchio, A., Cox, J. M., Eiermann, G. J., He, H., Lyons, K. A., Patel, R.

A., Patel, S. B., Petrov, A., Scapin, G., Wu, J. K., Xu, S., Zhu, B., Thornberry, N. A., Roy,

R. S., & Weber, A. E. (2009). Aminopiperidine-fused imidazoles as dipeptidyl peptidase-

IV inhibitors. Bioorganic & medicinal chemistry letters, 19(15), 4097–4101.

Eitsuka, T., Tatewaki, N., Nishida, H., Nakagawa, K., Miyazawa, T. (2016). Synergist ic

Anticancer Effect of Tocotrienol Combined with Chemotherapeutic Agents or Dietary

Components. International Journal of Molecular Sciences, 17(10), 1605.

Esposito, S., Bianco, A., Russo, R., Di Maro, A., Isernia, C., Pedone, P. V. (2019). Therapeutic

Perspectives of Molecules from Urtica dioica Extracts for Cancer Treatment. Molecules

(Basel, Switzerland), 24(15), 2753.

Falzone, L., Salomone, S., & Libra, M. (2018). Evolution of Cancer Pharmacological Treatments

at the Turn of the Third Millennium. Frontiers in pharmacology, 9, 1300.

Farkhondeh, T., Samarghandian, S., Borji, A. (2016). Evaluation of Antidiabetic Activity of

Carnosol (Phenolic Diterpene in Rosemary) in Streptozotocin-Induced Diabetic Rats.

Cardiovascular & hematological disorders drug targets, 17 (1), 11-17.

FILTER 2.5.1.4 (2013) OpenEye Scientific Software, Santa Fe, NM, http://www.eyesopen.co

Fletcher, S. (2019). MCL-1 inhibitors - where are we now (2019)?. Expert opinion on

therapeutic patents, 29(11), 909–919.

Page 124: Computational Approach Towards Exploring the

415

Folkerd, E. J., & Dowsett, M. (2010). Influence of sex hormones on cancer progression. Journal

of clinical oncology : official journal of the American Society of Clinical Oncology,

28(26), 4038–4044.

FRED 3.2.0.2 (2015) OpenEye Scientific Software, Santa Fe, NM, http://www.eyesopen.com

Fruman, D., Chiu, H., Hopkins, B., Bagrodia, S., Cantley, L., Abraham, R. (2017). The PI3K

Pathway in Human Disease. Cell, 170, 605-635.

Fu, H. Y., Chen, S. J., Chen, R. F., Ding, W. H., Kuo-Huang, L. L., Huang, R. N. (2006).

Identification of oxalic acid and tartaric acid as major persistent pain-inducing toxins in the

stinging hairs of the nettle, Urtica thunbergiana. Annals of botany, 98(1), 57–65 .

Fu, J., Chen, D., Zhao, B., Zhao, Z., Zhou, J., Xu, Y., Xin, Y., Liu, C., Luo, L., & Yin, Z. (2012).

Luteolin induces carcinoma cell apoptosis through binding Hsp90 to suppress constitutive

activation of STAT3. PloS one, 7(11), 49194.

Gaboriaud-Kolar, N., & Skaltsounis, A. L. (2013). Glycogen phosphorylase inhibitors: a patent

review (2008 - 2012). Expert opinion on therapeutic patents, 23(8), 1017–1032.

Gadewal, N. & Varma, A. (2012). Targeting Pim-1 kinase for potential drug-development.

International journal of computational biology and drug design. 5. 137-51.

Ganesan, A. & Barakat, K. (2016). Target fishing: a key to unlock the One-To-Many puzzle in

drug discovery. Journal of Pharmaceutical Care & Health Systems, 3(2),141.

Ganeshpurkar, A., & Saluja, A. K. (2017). The Pharmacological Potential of Rutin. Saudi

pharmaceutical journal : SPJ : the official publication of the Saudi Pharmaceutical Society,

25(2), 149–164.

Gao, Y., Liu, F., Fang, L., Cai, R., Zong, C. (2014). Genkwanin Inhibits Proinflammatory

Mediators Mainly through the Regulation of miR-101/MKP-1/MAPK Pathway in LPS-

Activated Macrophages. PLoS ONE, 9(5), 96741.

García-Reyes, B., Kretz, A. L., Ruff, J. P., von Karstedt, S., Hillenbrand, A., Knippschild, U.,

Henne-Bruns, D., & Lemke, J. (2018). The Emerging Role of Cyclin-Dependent Kinases

(CDKs) in Pancreatic Ductal Adenocarcinoma. International journal of molecular sciences,

19(10), 3219.

Gerber, D. E. (2008). EGFR Inhibition in the Treatment of Non-Small Cell Lung Cancer. Drug

development research, 69(6), 359–372.

Gfeller D., Michielin O. and Zoete V., (2013). "Shaping the interaction landscape of bioactive

molecules". Bioinformatics. 29:3073–3079

Page 125: Computational Approach Towards Exploring the

416

Gfeller, D., Grosdidier, A., Wirth, M., Daina, A., Michielin, O., & Zoete, V. (2014).

SwissTargetPrediction: a web server for target prediction of bioactive small molecules.

Nucleic Acids Research, 42(1), 32–38.

Ghanbari, R., Anwar, F., Alkharfy, K. M., Gilani, A. H., Saari, N. (2012). Valuable nutrients and

functional bioactives in different parts of olive (Olea europaea L.). International journal of

molecular sciences, 13(3), 3291–3340 .

Ghorbani, A. & Esmaeilizadeh, M. (2017). Pharmacological properties of Salvia officinalis and

its components. Journal of traditional and complementary medicine, 7(4), 433–440.

Giuseppe, M., Alessio, L., Marco, Mor., Riccardo, C. (2019). Fibroblast growth factor receptor

inhibitors: patent review (2015-2019), Expert Opinion onTherapeutic Patent, 29(12), 965-

977.

Gotto, A. M. & Moon, J. (2010). Pitavastatin for the treatment of primary hyperlipidemia and

mixed dyslipidemia. Expert review of cardiovascular therapy, 8(8), 1079–1090.

Govindaraj, J., & Sorimuthu, P. S. (2015). Rosmarinic acid modulates the antioxidant status and

protects pancreatic tissues from glucolipotoxicity mediated oxidative stress in high-fat diet:

streptozotocin-induced diabetic rats. Molecular and cellular biochemistry, 404(1-2), 143–

159 .

Grauso, L., de Falco, B., Lanzotti, V., Motti, R. (2020). Stinging nettle, Urtica dioica L.:

botanical, phytochemical and pharmacological overview. Journal of Phytochemistry

Reviews, 1-37.

Gül, S., Demirci, B., Başer, K. H., Akpulat, H. A., & Aksu, P. (2012). Chemical composition and

in vitro cytotoxic, genotoxic effects of essential oil from Urtica dioica L. Bulletin of

environmental contamination and toxicology, 88(5), 666–671.

Gülçin, I., Küfrevioglu, O. I., Oktay, M., Büyükokuroglu, M.E. (2004). Antioxidant,

antimicrobial, antiulcer and analgesic activities of nettle (Urtica dioica L.). Journal of

ethnopharmacology, 90(2-3), 205–215.

Guo, Y., Huang, Z., Sang, D., Gao, Q., & Li, Q. (2020). The Role of Nutrition in the Prevention

and Intervention of Type 2 Diabetes. Frontiers in bioengineering and biotechnology, 8,

575442.

Gupta, A. K., Sharma, S., Dahiya, N., Brashier, D. B. (2016). Palbociclib: A breakthrough in

breast carcinoma in women. Medical journal, Armed Forces India, 72(1), 37–42 .

Haikarainen, T., Krauss, S. & Lehtio, L. (2014). Tankyrases: structure, function and therapeutic

implications in cancer. Current pharmaceutical design, 20(41), 6472–6488.

Page 126: Computational Approach Towards Exploring the

417

Hajhashemi, V., & Klooshani, V. (2013). Antinociceptive and anti-inflammatory effects of

Urtica dioica leaf extract in animal models. Avicenna journal of phytomedicine, 3(2), 193–

200.

Hallinan, N., Finn, S., Cuffe, S., Rafee, S., O'Byrne, K., & Gately, K. (2016). Targeting the

fibroblast growth factor receptor family in cancer. Cancer treatment reviews, 46, 51–62.

Halliwell, B. (2007). Oxidative stress and cancer: Have we moved forward? Biochemical

Journal, 401(1), 1–11.

Han, J., Wang, D., Ye, L., Li, P., Hao, W., Chen, X., Ma, J., Wang, B., Shang, J., Li, D., Zheng,

Q. (2017). Rosmarinic Acid Protects against Inflammation and Cardiomyocyte Apoptosis

during Myocardial Ischemia/Reperfusion Injury by Activating Peroxisome Proliferator-

Activated Receptor Gamma. Frontiers in pharmacology, 8, 456.

Hawkins, P.C.D., Skillman, A.G., Nicholls, A. (2007). Comparison of Shape-Matching and

Docking as Virtual Screening Tools, Journal of Medicinal Chemistry, 50, 74-82.

He, J.H., Chen, L.X., Li, H.(2019). Progress in the discovery of naturally occurring anti-diabetic

drugs and in the identification of their molecular targets. Fitoterapia, 134, 270-289.

Hilborn, E., Stål, O., & Jansson, A. (2017). Estrogen and androgen-converting enzymes 17β-

hydroxysteroid dehydrogenase and their involvement in cancer: with a special focus on

17β-hydroxysteroid dehydrogenase type 1, 2, and breast cancer. Oncotarget, 8(18), 30552–

30562.

Himmat, M., Salim, N., Al-Dabbagh, M.M., Saeed, F., Ahmed, A. (2016). Adapting Document

Similarity Measures for Ligand-Based Virtual Screening. Molecules (Basel), 21(4), 476.

Ho, S. T., Tung, Y. T., Kuo, Y. H., Lin, C. C., & Wu, J. H. (2015). Ferruginol inhibits non-small

cell lung cancer growth by inducing caspase-associated apoptosis. Integrative cancer

therapies, 14(1), 86–97.

Hodroj, M. H., Al Bast, N., Taleb, R. I., Borjac, J., & Rizk, S. (2020). Nettle Tea Inhibits Growth

of Acute Myeloid Leukemia Cells In Vitro by Promoting Apoptosis. Nutrients, 12(9),

2629.

Hosseinimehr, S. J. (2014).A review of preventive and therapeutic effects of curcumin in patients

with cancer. Clinical Excellence, 2(2), 50-63.

Hoter, A., El-Sabban, M. E. & Naim, H. Y. (2018). The HSP90 Family: Structure, Regulation,

Function, and Implications in Health and Disease. International journal of molecular

sciences, 19(9), 2560.

Hsu, L. H., Chu, N. M. & Kao, S. H. (2017). Estrogen, Estrogen Receptor and Lung Cancer.

International journal of molecular sciences, 18(8), 1713.

Page 127: Computational Approach Towards Exploring the

418

Hua, H., Zhang, H., Kong, Q., & Jiang, Y. (2018). Mechanisms for estrogen receptor expression

in human cancer. Experimental hematology & oncology, 7, 24.

Huang, S.M., Mishina, Y.M., Liu, S., Cheung, A., Stegmeier, F., Michaud, G.A., Charlat, O.,

Wiellette, E., Zhang, Y., Wiessner, S. (2009).Tankyrase inhibition stabilizes axin and

antagonizes Wnt signalling. Nature, 461, 614-620.

Huang, Y., Chen, L., Guo, L., Hupp, T.R., Lin, Y. (2014). Evaluating DAPK as a therapeutic

target. Apoptosis, 19(2), 371-86.

Ibrahim, M., Rehman, K., Razzaq, A., Hussain, I., Farooq, T., Qadir, M., Akash, M. (2018).

Investigations of phytochemical constituents and their pharmacological properties isolated

from genus utrica: Critical review and analysis. Critical Reviews in Eukaryotic Gene

Expression, 28(1), 25-66.

Imran, M., Ahmad, N., Anjum, F. M., Khan, M. K., Mushtaq, Z., Nadeem, M., Hussain, S.

(2015). Potential protective properties of flax lignan secoisolariciresinol diglucoside.

Nutrition journal, 14, 71 .

International Agency for Research on Cancer, IARC, 12 September 2018

International Diabetes Federation (IDF) (2015) Key findings 2014, diabetes in the regions.

Retrieved from http://www.diabetesatlas.org/key-messages.html. Accessed 17 Mar 2018

International Diabetes Federation IDF, Diabetes Atlas Ninth edition 2019.

Jakovljević, M., Jokić, S., Molnar, M., Jašić, M., Babić, J., Jukić, H., Banjari, I. (2019).

Bioactive Profile of Various Salvia officinalis L. Preparations. Plants (Basel), 8(3), 55.

Jameera, B. A., Jubie, S., & Nanjan, M. J. (2017). Estrogen receptor agonists/antagonists in

breast cancer therapy: A critical review. Bioorganic chemistry, 71, 257–274 .

Jedinák, A., Mucková, M., Kost'álová, D., Maliar, T., Masterova, I. (2006). Antiprotease and

antimetastatic activity of ursolic acid isolated from Salvia officinalis. Journal of

biosciences, 61(11-12), 777–782.

Jeon, G.A., Lee, J.S., Patel, V., Gutkind, J.S., Thorgeirsson, S.S., Kim, E.C., Chu, I.S.,

Amornphimoltham, P., Park, M.H. (2004). Global gene expression profiles of human head

and neck squamous carcinoma cell lines. International Journal of Cancer, 112, 249–258.

Jian, Y., He, Y., Yang, J., Han, W., Zhai, X., Zhao, Y., & Li, Y. (2018). Molecular Modeling

Study for the Design of Novel Peroxisome Proliferator-Activated Receptor Gamma

Agonists using 3D-QSAR and Molecular Docking. International journal of molecular

sciences, 19(2), 630.

Jiang, Y., Zhang, L., & Rupasinghe, V. (2016).The anticancer properties of phytochemical

extracts from Salvia plants. Botanics: Targets and Therapy, 6, 25-44.

Page 128: Computational Approach Towards Exploring the

419

John, S. 2020. Senhwa Announces Multiple IND application submissions to U.S. FDA for

Silmitasertib as a Potential Treatment for COVID-19. Senhwa Biosciences.

Johnson, J. J. (2011). Carnosol: a promising anti-cancer and anti-inflammatory agent. Cancer

letters, 305(1), 1–7.

Jordan, V. C. (2007). Beyond raloxifene for the prevention of osteoporosis and breast cancer.

British journal of pharmacology, 150(1), 3–4.

Jorgensen, W.L. (2010). Drug discovery: Pulled from a protein‘s embrace. Nature, 466, 42–43.

Joshi, B., Mukhija, M. & Kalia, A. (2014). Pharmacognostical review of Urtica dioica L..

International Journal of Green Pharmacy, 8(4), 201-209.

Kar, Ashutosh. (2006) . Pharmacognosy and Pharmacobiotechnology. second edition. 5-11

Kataki, M., Murugamani, V., Rajkumari, A., Mehra, P., Awasthi, D., Yadav, R. (2012).

Antioxidant, hepatoprotective and anthelmintic activities of methanol extract of Urtica

dioica L. Leaves. Pharm Crops, 3, 38-46.

Katiyar, C., Gupta, A., Kanjilal, S., Katiyar, S. (2012). Drug discovery from plant sources: An

integrated approach. Ayu, 33(1), 10–19.

Kearnes S. & Pande V., (2016). "ROCS-derived features for virtual screening". Journal of

Comput Aided Molecular Design, 30(8), 609-617.

Keeton, E., Mcachern, K., Alimzhanov, M., Wang, S., Cao, Y., Bao, L., Palakurthi, S.,

Grondine, M., Chen, Y., Dillman, K. (2012). Abstract 2796: Efficacy and biomarker

modulation by AZD1208, a novel, potent and selective pan-Pim kinase inhibitor, in models

of acute myeloid leukemia. Cancer Research, 72(8), 2796-2796.

Khalife, R., Hodroj, M. H., Fakhoury, R., Rizk, S. (2016). Thymoquinone from Nigella sativa

Seeds Promotes the Antitumor Activity of Noncytotoxic Doses of Topotecan in Human

Colorectal Cancer Cells in Vitro. Planta medica, 82(4), 312–321.

Kianbakht, S., Khalighi-Sigaroodi, F., & Dabaghian, F. H. (2013). Improved glycemic control in

patients with advanced type 2 diabetes mellitus taking Urtica dioica leaf extract: a

randomized double-blind placebo-controlled clinical trial. Clinical laboratory, 59(9-10),

1071–1076.

Kim, G. J., Jo, H. J., Lee, K. J., Choi, J. W., & An, J. H. (2018). Oleanolic acid induces p53-

dependent apoptosis via the ERK/JNK/AKT pathway in cancer cell lines in prostatic

cancer xenografts in mice. Oncotarget, 9(41), 26370–26386.

Kinnings, S. L., Liu, N., Buchmeier, N., Tonge, P. J., Xie, L., & Bourne, P. E. (2009). Drug

discovery using chemical systems biology: repositioning the safe medicine Comtan to treat

Page 129: Computational Approach Towards Exploring the

441

multi-drug and extensively drug resistant tuberculosis. PLoS computational biology, 5(7),

1000423.

Kinoshita, T., Nakaniwa, T., Sekiguchi, Y., Sogabe, Y., Sakurai, A., Nakamura, S., & Nakanishi,

I. (2013). Crystal structure of human CK2α at 1.06 Å resolution. Journal of synchrotron

radiation, 20(6), 974–979.

Kıyga, E., Şengelen, A., Adıgüzel, Z., Önay Uçar, E. (2020). Investigation of the role of

quercetin as a heat shock protein inhibitor on apoptosis in human breast cancer cells.

Molecular Biology Reports, 47(7):4957-4967.

Kondeva-Burdina, M., Krasteva, I., & Mitcheva, M. (2014). Effects of rhamnocitrin 4-β-D-

galactopyranoside, isolated from Astragalus hamosus on toxicity models in vitro.

Pharmacognosy magazine, 10(3), 487–493.

Kontogianni, V., Tomic, G., Nikolic, I., Nerantzaki, A., Sayyad, N., Stosic-Grujicic, S.,

Stojanovic, I., Gerothanassis, I., Tzakos, A. (2013). Phytochemical profile of Rosmarinus

officinalis and Salvia officinalis extracts and correlation to their antioxidant and anti-

proliferative activity. Food chemistry,136,120-9.

Kooti, W., Servatyari, K., Behzadifar, M., Asadi-Samani, M., Sadeghi, F., Nouri, B., Zare-

Marzouni, H. (2017). Effective Medicinal Plant in Cancer Treatment, Part 2: Review

Study. Journal of Evidence-Based Complementary & Alternative Medicine, 22(4), 982–

995 .

Koparde, A. A., Chandrashekar, R., Chandrakant, S. M. (2019). Natural Products in Drug

Discovery, Pharmacognosy - Medicinal Plants.

Kumar, A., Mandiyan, V., Suzuki, Y., Zhang, C., Rice, J., Tsai, J., Artis, D.R., Ibrahim, P. and

Bremer, R. (2005) ‗Crystal structures of proto-oncogene kinase Pim1: a target of aberrant

somatic hypermutations in diffuse large cell lymphoma‘, Journal of Molecular Biology,

348, 183–193.

Kumar, S., & Pandey, A. K. (2013). Chemistry and biological activities of flavonoids: an

overview. The Scientific World Journal, 2013, 162750.

Lau, T., Chan, E., Callow, M., Waaler, J., Boggs, J., Blake, R. A., Magnuson, S., Sambrone, A.,

Schutten, M., Firestein, R., Machon, O., Korinek, V., Choo, E., Diaz, D., Merchant, M.,

Polakis, P., Holsworth, D. D., Krauss, S., & Costa, M. (2013). A novel tankyrase small-

molecule inhibitor suppresses APC mutation-driven colorectal tumor growth. Cancer

research, 73(10), 3132–3144.

Le, B. T., Kumarasiri, M., Adams, J. R., Yu, M., Milne, R., Sykes, M. J., Wang, S. (2015).

Targeting Pim kinases for cancer treatment: opportunities and challenges. Future medicinal

chemistry, 7(1), 35–53.

Page 130: Computational Approach Towards Exploring the

444

Lee, J., Park, T., Jeong, S., Kim, K. H., Hong, C. (2007). 3-Hydroxychromones as cyclin-

dependent kinase inhibitors: synthesis and biological evaluation. Bioorganic & medicinal

chemistry letters, 17(5), 1284–1287.

Lee, S. Y., Kim, Y. J., Chung, S. O., Park, S. U. (2016). Recent studies on ursolic acid and its

biological and pharmacological activity. Excli journal, 15, 221–228.

Leelananda, S. P., & Lindert, S. (2016). Computational methods in drug discovery. Beilstein

journal of organic chemistry, 12, 2694–2718.

Li, T., Zhou, W., Bi, H., Zhuang, Y., Zhang, T., & Liu, T. (2018). Production of caffeoylmalic

acid from glucose in engineered Escherichia coli. Biotechnology Letters, 40(7), 1057–

1065.

Liang, W., Wu, X., Fang, W., Zhao, Y., Yang, Y., Hu, Z., Xue, C., Zhang, J., Zhang, J., Ma, Y.,

Zhou, T., Yan, Y., Hou, X., Qin, T., Dinglin, X., Tian, Y., Huang, P., Huang, Y., Zhao, H.,

& Zhang, L. (2014). Network meta-analysis of erlotinib, gefitinib, afatinib and icotinib in

patients with advanced non-small-cell lung cancer harboring EGFR mutations. PloS one,

9(2), 85245.

Litchfield, D. W. (2003). Protein kinase CK2: structure, regulation and role in cellular decisions

of life and death. The Biochemical journal, 369(1), 1–15.

Liu, D., Gao, H., Tang, W. (2017). Plant non-starch polysaccharides that inhibit key enzymes

linked to type 2 diabetes mellitus. Annals of the New York Academy of Sciences, 1401,

(1), 28-36.

Liu, J., Cao, X., Xiao, Q., Quan, M. (2015). Apigenin inhibits HeLa sphere-forming cells

through inactivation of casein kinase 2α. Molecular Medicine Reports, 11, 665-669.

Liu, P., Cheng, H., Roberts, T. M., Zhao, J. J. (2009). Targeting the phosphoinositide 3-kinase

pathway in cancer. Nature reviews. Drug discovery, 8(8), 627–644.

Liu, Z., Fang, H., Reagan, K., Xu, X., Mendrick, D. L., Slikker, W., Jr, & Tong, W. (2013). In

silico drug repositioning: what we need to know. Drug discovery today, 18(3-4), 110–115.

Lo, Y. C., Senese, S., Damoiseaux, R., & Torres, J. Z. (2016). 3D Chemical Similarity Networks

for Structure-Based Target Prediction and Scaffold Hopping. ACS chemical biology,

11(8), 2244–2253.

Lolli, G., Cozza, G., Mazzorana, M., Tibaldi, E., Cesaro, L., Donella-Deana, A., Meggio, F.,

Venerando, A., Franchin, C., Sarno, S., Battistutta, R., & Pinna, L. A. (2012). Inhibition of

protein kinase CK2 by flavonoids and tyrphostins. A structural insight. Biochemistry,

51(31), 6097–6107.

Luo, G., Zhou, J., Li, G., Hu, N., Xia, X., & Zhou, H. (2019). Ferruginol Diterpenoid Selectively

Inhibits Human Thyroid Cancer Growth by Inducing Mitochondrial Dependent Apoptosis,

Page 131: Computational Approach Towards Exploring the

442

Endogenous Reactive Oxygen Species (ROS) Production, Mitochondrial Membrane

Potential Loss and Suppression of Mitogen-Activated Protein Kinase (MAPK) and

PI3K/AKT Signaling Pathways. Medical science monitor. International medical journal of

experimental and clinical research, 25, 2935–2942 .

Luszczak, S., Kumar, C., Sathyadevan, V. K., Simpson, B. S., Gately, K. A., Whitaker, H. C.,

Heavey, S. (2020). PIM kinase inhibition: co-targeted therapeutic approaches in prostate

cancer. Signal Transduction and Targeted Therapy, 5(1), 7.

Mahmood, A. & Alkhathlan, H. Z. (2019). Isolation, synthesis and pharmacological applications

of cirsimaritin – A short review. Journal of Medicinal Plants, 7(11), 252-260.

Malinowsky, K., Wolff, C., Gündisch, S., Berg, D., & Becker, K. (2015). Targeted therapies in

cancer - challenges and chances offered by newly developed techniques for protein

analysis in clinical tissues. Journal of Cancer, 2, 26–35.

Mangione, W. & Samudrala, R. (2019). Identifying Protein Features Responsible for Improved

Drug Repurposing Accuracies Using the CANDO Platform: Implications for Drug Design.

Molecules (Basel, Switzerland), 24(1), 167.

Markham A. (2019). Erdafitinib: First Global Approval. Drugs, 79(9), 1017–1021.

Maruthanila, V. L., Elancheran, R., Roy, N. K., Bhattacharya, A., Kunnumakkara, A. B.,

Kabilan, S., Kotoky, J. (2019). In silico Molecular Modelling of Selected Natural Ligands

and their Binding Features with Estrogen Receptor Alpha. Current computer-aided drug

design, 15(1), 89–96.

Masłyk, M., Janeczko, M., Martyna, A., Kubiński, K. (2017). CX-4945: The protein kinase CK2

inhibitor and anti-cancer drug shows anti-fungal activity. Molecular and cellular

biochemistry, 435(1-2), 193–196.

Mau, S., Saleem, S., Kimothi, V., Joshi, V., Singh, S. (2019). An exhaustive review on herbs

used in the management of diabetes mellitus type 2. Journal of Applied Pharmaceutical

Sciences and Research, 2(2), 5-11.

Medina, J. R., Blackledge, C. W., Heerding, D. A., Campobasso, N., Ward, P., Briand, J.,

Wright, L., Axten, J. M. (2010). Aminoindazole PDK1 Inhibitors: A Case Study in

Fragment-Based Drug Discovery. ACS medicinal chemistry letters, 1(8), 439–442.

Miroddi, M., Navarra, M., Quattropani, M. C., Calapai, F., Gangemi, S., & Calapai, G. (2014).

Systematic review of clinical trials assessing pharmacological properties of Salvia species

on memory, cognitive impairment and Alzheimer's disease. CNS neuroscience &

therapeutics, 20(6), 485–495.

Miura, T., Takagi, S., & Ishida, T. (2012). Management of Diabetes and Its Complications with

Banaba (Lagerstroemia speciosa L.) and Corosolic Acid. Evidence-based complementary

and alternative medicine, 2012(5), 871495.

Page 132: Computational Approach Towards Exploring the

442

Mohammadi, A., Mansoori, B., Aghapour, M., & Baradaran, B. (2016). Urtica dioica

dichloromethane extract induce apoptosis from intrinsic pathway on human prostate cancer

cells (PC3). Cellular and molecular biology (Noisy-le-Grand, France), 62(3), 78–83.

Mohammed, Z., Nada, S., Al-Halbosiy, M., Abdulfattah, Sh., Abdul-Hameed, B. (2014).

Cytotoxic effects of Ammi visnaga volatile oil on some cancer cell lines. Jornal of

Biotechnology Research Center,8(1), 5-7.

Mohan, N., & Latha, M.S., (2018). Insilico Docking And Interaction Analysis Of Ellagic Acid

And Curcumin Derivatives Against Human Cancer. Indian J.Sci.Res. 18(2): 22-28.

Mondello, P., Cuzzocrea, S., & Mian, M. (2014). Pim kinases in hematological malignancies:

where are we now and where are we going?. Journal of hematology & oncology, 7, 95.

Mora, A., Komander, D., van Aalten, D. M. F., & Alessi, D. R. (2004). PDK1, the master

regulator of AGC kinase signal transduction. Seminars in Cell & Developmental Biology,

15(2), 161–170.

Morris, A. P., Voight, B. F., Teslovich, T. M., Ferreira, T., Segrè, A. V., Steinthorsdottir, V.,

Strawbridge, R. J., Khan, H., Grallert, H., Mahajan, A., Prokopenko, I., Kang, H. M., Dina,

C., Esko, T., Fraser, R. M., Kanoni, S., Kumar, A., Lagou, V., Langenberg, C., (2012).

Diabetes Genetics Replication And Meta-analysis (DIAGRAM) Consortium. Large-scale

association analysis provides insights into the genetic architecture and pathophysiology of

type 2 diabetes. Nature genetics, 44(9), 981–990.

Mueen, A., & Parasuraman, S. (2016). Urtica dioica L., (Urticaceae): A Stinging Nettle.

Systematic Reviews in Pharmacy. 5(1), 6-8.

Nagashima, K., Shumway, S. D., Sathyanarayanan, S., Chen, A. H., Dolinski, B., Xu, Y.,

Keilhack, H., Nguyen, T., Wiznerowicz, M., Li, L., Lutterbach, B. A., Chi, A., Paweletz,

C., Allison, T., Yan, Y., Munshi, S. K., Klippel, A., Kraus, M., Bobkova, E. V.,

Deshmukh, S., … Andersen, J. N. (2011). Genetic and pharmacological inhibition of

PDK1 in cancer cells: characterization of a selective allosteric kinase inhibitor. The Journal

of biological chemistry, 286(8), 6433–6448.

Nagy, L., Docsa, T., Szántó, M., Brunyánszki, A., Hegedűs, C., Márton, J., Kónya, B., Virág, L.,

Somsák, L., Gergely, P., & Bai, P. (2013). Glycogen phosphorylase inhibitor N-(3,5-

dimethyl-Benzoyl)-N'-(β-D-glucopyranosyl)urea improves glucose tolerance under

normoglycemic and diabetic conditions and rearranges hepatic metabolism. PloS one, 8(7),

69420.

Nagy, L., Márton, J., Vida, A., Kis, G., Bokor, É., Kun, S., Gönczi, M., Docsa, T., Tóth, A.,

Antal, M., Gergely, P., Csóka, B., Pacher, P., Somsák, L., & Bai, P. (2018). Glycogen

phosphorylase inhibition improves beta cell function. British journal of pharmacology,

175(2), 301–319.

Page 133: Computational Approach Towards Exploring the

441

Nahata, A., & Dixit, V. K. (2012). Ameliorative effects of stinging nettle (Urtica dioica) on

testosterone-induced prostatic hyperplasia in rats. Andrologia, 44(1), 396–409.

Narwal, M., Koivunen, J., Haikarainen, T., Obaji, E., Legala, O., Venkannagari, H., Joensuu, P.,

Pihlajaniemi, T., Lehtiö, L. (2013). Discovery of Tankyrase Inhibiting Flavones with

Increased Potency and Isoenzyme Selectivity. Journal of Medicinal Chemistry, 56(20),

7880–7889.

Nath Choudhury, P. (2014). A Study of in-silico Docking and ADME/Tox Property of Known

Flavonoid Molecule Hispidulin. Pursuits, 5, 34-40.

National Cancer Institute. (2015a). What Is Cancer? https://www.cancer.gov/aboutcancer/

understanding/what-is-cancer

National Cancer Institute. (2015b). Research on Causes of Cancer https://www.cancer.gov/

research/areas/causes

Naveed, M., Hejazi, V., Abbas, M., Kamboh, A. A., Khan, G. J., Shumzaid, M., Ahmad, F.,

Babazadeh, D., FangFang, X., Modarresi-Ghazani, F., WenHua, L., & XiaoHui, Z. (2018).

Chlorogenic acid (CGA): A pharmacological review and call for further research.

Biomedicine & pharmacotherapy, 97, 67–74.

Navid, S., Fan, C., O. Flores-Villanueva, P., Generali, D., & Li, Y. (2020). The Fibroblast

Growth Factor Receptors in Breast Cancer: from Oncogenesis to Better Treatments.

International Journal of Molecular Sciences, 21(6), 2011.

Nawab, A., Yunus, M., Mahdi, A., & Gupta, S. (2011). Evaluation of Anticancer Properties of

Medicinal Plants from the Indian Sub-Continent. Molecular and Cellular Pharmacology, 3,

21-29.

Nedunuri, D., Adimulam, Y. b. & Reddi, K. K. (2016). In silico pathway analysis of MAPKs and

computational investigation of bipyrazole analogues as novel p38alpha MAPK inhibitors.

International Journal of Bioinformatics Research and Applications, 12(1), 1.

Nettles, J., Jenkins, J., Bender, A., Deng, Z., Davies, J. W., & Glick, M. (2006). Bridging

chemical and biological space: "target fishing" using 2D and 3D molecular descriptors.

Journal of medicinal chemistry, 49(23), 6802–6810.

Newman, D. J., & Cragg, G. M. (2007). Natural products as sources of new drugs over the last

25 years. Journal of natural products, 70(3), 461–477.

Newman, D. J., & Cragg, G. M. (2016). Natural Products as Sources of New Drugs from 1981 to

2014. Journal of natural products, 79(3), 629–661.

OEDOCKING3.2.0.2,OpenEye Scientific Software, Santa Fe, NM. (http://www.

Eyesopen.com), (2015).

Page 134: Computational Approach Towards Exploring the

445

OMEGA 2.5.1.4. (2013) OpenEye Scintific Software, Santa Fe, NM, http://www.eyesopen.com.

Ornitz, M., & Itoh, N. (2015). The Fibroblast Growth Factor signaling pathway. Wiley

interdisciplinary reviews. Developmental biology, 4(3), 215–266.

Ortega, C. E., Seidner, Y., & Dominguez, I. (2014). Mining CK2 in cancer. PloS one, 9(12),

115609.

Osmi S., Begić, S., Mićic, V., Petrovic, Z., Avdić., G. (2019). Effect of solvent and extraction

conditions on antioxidative activity of Sage (Salvia Officinalis L.) Extracts Obtained By

Maceration. Technologica Acta, 11,(2),1–8.

Otles, S., & Yalcin, B. (2012). Phenolic compounds analysis of root, stalk, and leaves of nettle.

TheScientificWorldJournal, 2012, 564367.

Padgett, C. S., Lim, C., Marschke, R. F., Northfelt, D. W., Andreopoulou, E., Hoff, V., O‘Brien,

S. (2010). 414 Clinical pharmacokinetics and pharmacodynamics of CX-4945, a novel

inhibitor of protein kinase CK2: Interim report from the phase 1 clinical trial. European

Journal of Cancer Supplements, 8(7), 131–132.

Padhye, S., Banerjee, S., Ahmad, A., Mohammad, R., & Sarkar, F. H. (2008). From here to

eternity - the secret of Pharaohs: Therapeutic potential of black cumin seeds and beyond.

Cancer therapy, 6(b), 495–510.

Pai, A., Lobo, R., Basu-Mallik, S., Jayashree, B. (2017). Extra precision docking studies of novel

luteolin analogues for the inhibition of Tankyrase II: A theoretical-based approach toward

novel cancer target. Thai Journal of Pharmaceutical Sciences, 41, 138-143.

Palestenian Ministry of Health Annual report (PMoH). (2016). The Palestinian Health

Information Center, Ministry of Health, Palestine.

Pang, X., Fu, W., Wang, J., Kang, D., Xu, L., Zhao, Y., Liu, A. L., & Du, G. H. (2018).

Identification of Estrogen Receptor α Antagonists from Natural Products via In Vitro and

In Silico Approaches. Oxidative medicine and cellular longevity, 2018, 6040149.

Papapetropoulos, A. & Szabo, C. (2018). Inventing new therapies without reinventing the wheel:

the power of drug repurposing. British journal of pharmacology, 175(2), 165–167.

Paramaguru, R., Mazumder, P. M., Sasmal, D., & Jayaprakash, V. (2014). Antidiabetic activity

of Pterospermum acerifolium flowers and glucose uptake potential of bioactive fraction in

L6 muscle cell lines with its HPLC fingerprint. BioMed research international, 2014(3),

459376.

Pares, L., Nonell-Canals, A., Sanchez-Martinez, M., & Avila, C. (2017). Computer-Aided Drug

Design Applied to Marine Drug Discovery: Meridianins as Alzheimer's Disease

Therapeutic Agents. Marine drugs, 15(12), 366.

Page 135: Computational Approach Towards Exploring the

446

Patel, K., & Patel, D. K. (2016). Medicinal importance, pharmacological activities, and

analytical aspects of hispidulin: A concise report. Journal of traditional and complementary

medicine, 7(3), 360–366.

Patterson, S., Wyllie, S., Norval, S., Stojanovski, L., Simeons, F. R., Auer, J. L., Osuna-Cabello,

M., Read, K. D., & Fairlamb, A. H. (2016). The anti-tubercular drug delamanid as a

potential oral treatment for visceral leishmaniasis. ELife, 5, 09744.

Pe, Onwe & Cs, Anyigor-Ogah & Okorocha, Albert & Ao, Afoke. (2015). Hyperlipidemia:

Etiology and Possible Control. Journal of Dental and Medical Sciences, 14, 93-100.

Petersen, M. C., & Shulman, G. I. (2018). Mechanisms of Insulin Action and Insulin Resistance.

Physiological reviews, 98(4), 2133–2223.

Phukan, S. (2010). "GSK3b: role in therapeutic landscape and development of modulators".

British Journal of Pharmacology, 160:1–19.

Pierre, F., Stefan, E., Nédellec, A. S., Chevrel, M. C., Regan, C. F., Siddiqui-Jain, A., Macalino,

D., Streiner, N., Drygin, D., Haddach, M., O'Brien, S. E., Anderes, K., & Ryckman, D. M.

(2011). 7-(4H-1,2,4-Triazol-3-yl)benzo[c][2,6]naphthyridines: a novel class of Pim kinase

inhibitors with potent cell antiproliferative activity. Bioorganic & medicinal chemistry

letters, 21(22), 6687–6692.

Pinzi, L., & Rastelli, G. (2019). Molecular Docking: Shifting Paradigms in Drug Discovery.

International journal of molecular sciences, 20(18), 4331.

Pourahmadi, M., Jashni, K., Bagheri, M., Jahromi, A. (2014). The effect of hydro-alcoholic

extract of Urtica dioica Root on testes in adult rats. Life Science Journal ,11(5), 420–424.

Powers, C., & Setzer, W. (2015). A molecular docking study of phytochemical estrogen mimics

from dietary herbal supplements. In Silico Pharmacology, 3(1), 1-63.

Puhl, A. C., Bernardes, A., Silveira, R. L., Yuan, J., Campos, J. L., Saidemberg, D. M., Palma,

M. S., Cvoro, A., Ayers, S. D., Webb, P., Reinach, P. S., Skaf, M. S., & Polikarpov, I.

(2012). Mode of peroxisome proliferator-activated receptor γ activation by luteolin.

Molecular pharmacology, 81(6), 788–799.

Quackenbush, K., Bagby, S., Tai, W., Messersmith, W., Schreiber, A., Greene, J., Kim, J.,

Wang, G., Purkey, A., Pitts, T. (2016). The novel tankyrase inhibitor (AZ1366) enhances

irinotecan activity in tumors that exhibit elevated tankyrase and irinotecan resistance.

Oncotarget 7, 28273-28285.

Raimondi, C., & Falasca, M. (2011). Targeting PDK1 in cancer. Current medicinal chemistry,

18(18), 2763–2769.

Ramos, M. C., Genilloud, O., Reyes, F., Vicente, F. (2018). Drug Discovery from Natural

Products for Pancreatic Cancer. Advances in Pancreatic Cancer. 135-175

Page 136: Computational Approach Towards Exploring the

447

Rebello, R. J., Huglo, A. V., & Furic, L. (2018). PIM activity in tumours: A key node of therapy

resistance. Advances in biological regulation, 67, 163–169.

Reddy, A. & Zhang S. (2013). "Polypharmacology: drug discovery for the future". Expert

Review of Clinical Pharmacology, 6(1), 41-47.

Reuter, S., Gupta, S. C., Chaturvedi, M. M., & Aggarwal, B. B. (2010). Oxidative stress,

inflammation, and cancer: how are they linked? Free radical biology & medicine, 49(11),

1603–1616.

Rivlin, N., Brosh, R., Oren, M., Rotter, V. (2011). Mutations in the p53 Tumor Suppressor Gene:

Important Milestones at the Various Steps of Tumorigenesis. Genes & cancer, 2(4), 466–

474 .

ROCS 3.2.1.4. (2015) OpenEye Scintific Software, Santa Fe, NM, http://www.eyesopen.com

Rodgers, G. M., 3rd, Becker, P. S., Blinder, M., Cella, D., Chanan-Khan, A., Cleeland, C.,

Coccia, P. F., Djulbegovic, B., Gilreath, J. A., Kraut, E. H., Matulonis, U. A., Millenson,

M. M., Reinke, D., Rosenthal, J., Schwartz, R. N., Soff, G., Stein, R. S., Vlahovic, G., &

Weir, A. B., 3rd (2012). Cancer- and chemotherapy- induced anemia. Journal of the

National Comprehensive Cancer Network : JNCCN, 10(5), 628–653.

Salazar, J. R., Loza-Mejía, M. A., & Soto-Cabrera, D. (2020). Chemistry, Biological Activities

and In Silico Bioprospection of Sterols and Triterpenes from Mexican Columnar

Cactaceae. Molecules, 25(7), 1649.

Salehi, B., Venditti, M., Sharifi-Rad, D., Kręgiel, J., Durazzo,H. (2019) .The Therapeutic

potential of apigenin. International Journal of Molecular Sciences, 20, 1305-1331.

Sampei, M., Arai, M. A., & Ishibashi, M. (2018). Total syntheses of schizandriside, saracoside

and (±)-isolariciresinol with antioxidant activities. Journal of Natural Medicines, 72(3),

651–654.

Santen, R. J., Song, R. X., McPherson, R., Kumar, R., Adam, L., Jeng, M. H., & Yue, W. (2002).

The role of mitogen-activated protein (MAP) kinase in breast cancer. The Journal of

steroid biochemistry and molecular biology, 80(2), 239–256.

Scannell, J. W., Blanckley, A., Boldon, H., & Warrington, B. (2012). Diagnosing the decline in

pharmaceutical R&D efficiency. Nature reviews. Drug discovery, 11(3), 191–200.

Schenone, M., Dančík, V., Wagner, B. K., & Clemons, P. A. (2013). Target identification and

mechanism of action in chemical biology and drug discovery. Nature chemical biology,

9(4), 232–240.

Scott, R. A., Lagou, V., Welch, R. P., Wheeler, E., Montasser, M. E., Luan, J., Mägi, R.,

Strawbridge, R. J., Rehnberg, E., Gustafsson, S., Kanoni, S., Rasmussen-Torvik, L. J.,

Yengo, L., Lecoeur, C., Shungin, D., Sanna, S., Sidore, C., Johnson, P. C., Barroso, I.

Page 137: Computational Approach Towards Exploring the

448

(2012). Large-scale association analyses identify new loci influencing glycemic traits and

provide insight into the underlying biological pathways. Nature genetics, 44(9), 991–1005.

Sepide, M. (2016).Therapeutic effects of Urtica dioica L: A review study. Der Pharma

Chemica,8 (17):287-292.

Sepúlveda, L., Ascacio-Valdés, J., Rodriguez, R., Aguilera-Carbo, A. & Aguilar, C. (2012).

Ellagic Acid: Biological Properties and Biotechnological Development for Production

Processes. African Journal Of Biotechnology, 10(5315), 4518-4523.

Shariatzadeh, S., Hamta, A., Soleimani., M, Fallah Huseini H.& Samavat, S. (2014).The

Cytotoxic Effects of Silymarin on the 4T1 Cell Line Derived from BALB/c Mice

Mammary Tumors. J. Med. Plants, 13 (52), 55-65.

Sharma, Y., Fagan, J. & Schaefer, J. (2019). Ethnobotany, phytochemistry, cultivation and

medicinal properties of Garden sage (Salvia officinalis L.), 8(3), 3139-3148.

Shattat, G. (2014). A Review Article on Hyperlipidemia: Types, Treatments and New Drug

Targets. Biomedical and Pharmacology Journal, 7(2), 399-409.

Shiloh, R., Bialik, S., & Kimchi, A. (2014). The DAPK family: a structure-function analysis.

Apoptosis : an international journal on programmed cell death, 19(2), 286–297.

Shumaila, A., Maryam S. & Ayesha N. (2016). A Mini Review On Cancer And Anticancer

Drugs. . Indo American Journal Of Pharmaceutical Sciences Iajps, 03(11), 1383–1388.

Siddiqui-Jain, A., Drygin, D., Streiner, N., Chua, P., Pierre, F., O'Brien, S. E., Bliesath, J.,

Omori, M., Huser, N., Ho, C., Proffitt, C., Schwaebe, M. K., Ryckman, D. M., Rice, W.

G., & Anderes, K. (2010). CX-4945, an orally bioavailable selective inhibitor of protein

kinase CK2, inhibits prosurvival and angiogenic signaling and exhibits antitumor efficacy.

Cancer research, 70(24), 10288–10298.

Singh, R., & Nain S (2018). A Mini-Review on Hyperlipidemia: Common Clinical Problem.

Journal of Interventional Cardiology, 4, 3-10.

Singh, Salam & Deb, Chitta & Ahmed, Sharif & Yenisetti, Sarat & Konwar, Bolin. (2015).

Molecular docking simulation analysis of the interaction of dietary flavonols with heat

shock protein 90. Journal of Biomedical Research. 30.(1), 67-74.

Sliwoski, G., Kothiwale, S., Meiler, J., & Lowe, E. W., (2013). Computational methods in drug

discovery. Pharmacological reviews, 66(1), 334–395.

Song, H. L., Zhang, X., Wang, W. Z., Liu, R. H., Zhao, K., Liu, M. Y., Gong, W. M., & Ning, B.

(2018). Neuroprotective mechanisms of rutin for spinal cord injury through anti-oxidation

and anti-inflammation and inhibition of p38 mitogen activated protein kinase pathway.

Neural regeneration research, 13(1), 128–134.

Page 138: Computational Approach Towards Exploring the

449

Sosa, V., Moliné, T., Somoza, R., Paciucci, R., Kondoh, H., & LLeonart, M. E. (2013).

Oxidative stress and cancer: an overview. Ageing research reviews, 12(1), 376–390.

Sousa Silva, M., Gomes, R. A., Ferreira, A. E., Ponces Freire, A., & Cordeiro, C. (2013). The

glyoxalase pathway: the first hundred years... and beyond. The Biochemical journal,

453(1), 1–15.

Srinivasan, P., Vijayakumar, S., Kothandaraman, S., & Palani, M. (2018). Anti-diabetic activity

of quercetin extracted from Phyllanthus emblica L. fruit: In silico and in vivo approaches.

Journal of pharmaceutical analysis, 8(2), 109–118 .

Stratford, E. W., Daffinrud, J., Munthe, E., Castro, R., Waaler, J., Krauss, S., & Myklebost, O.

(2014). The tankyrase-specific inhibitor JW74 affects cell cycle progression and induces

apoptosis and differentiation in osteosarcoma cell lines. Cancer medicine, 3(1), 36–46.

Stravodimos, G. A., Kantsadi, A. L., Apostolou, A., Kyriakis, E., Kafaski-Kanelli, V. N.,

Solovou, T., Gatzona, P., Liggri, P. G. V., Theofanous, S., Gorgogietas, V. A., Kissa, A.,

Psachoula, C., Lemonakis, A., Chatzileontiadou, D. S. M., Psarra, A. M. G., Skamnaki, V.

T., Haroutounian, S. A., & Leonidas, D. D. (2018). Affinity crystallography reveals the

bioactive compounds of industrial juicing byproducts of Punica granatum for glycogen

phosphorylase. Current Drug Discovery Technologies, 15(1), 41-53.

Sun, J. (2016). PTP1B, A potential target of type 2 diabetes mellitus. Journal of Molecular

Biology. 5:(4):174-180.

Suvarna, V., Murahari, M., Khan, T., Chaubey, P., & Sangave, P. (2017). Phytochemicals and

PI3K Inhibitors in Cancer-An Insight. Frontiers in pharmacology, 8, 916.

Swargam, S., Pradhan , D. & Umamaheswari, A.(2010). Identification of Potent Leads for

Human cAMP Dependent Protein Kinase Catalytic Subunit Alpha: A Strategic Application

of Virtual Screening for Cancer Therapeutics. Nature Precedings. doi.org/ 10.1038/npre.

2010. 4903.1

Takasawa, R., Shimada, N., Uchiro, H., Takahashi, S., Yoshimori, A., & Tanuma, S. (2016).

TLSC702, a Novel Inhibitor of Human Glyoxalase I, Induces Apoptosis in Tumor Cells.

Biological & pharmaceutical bulletin, 39(5), 869–873.

Thomas, L. W., Lam, C., & Edwards, S. W. (2010). Mcl-1; the molecular regulation of protein

function. Federation of European Biochemical Societies (FEBS) Letters, 584(14), 2981–

2989.

Thomson, M., & Ali, M. (2003). Garlic [Allium sativum]: a review of its potential use as an anti-

cancer agent. Current cancer drug targets, 3(1), 67–81.

Thornalley, P. J., & Rabbani, N. (2011). Glyoxalase in tumourigenesis and multidrug resistance.

Seminars in cell & developmental biology, 22(3), 318–325.

Page 139: Computational Approach Towards Exploring the

421

Tian, X. H., Hou, W. J., Fang, Y., Fan, J., Tong, H., Bai, S. L., Chen, Q., Xu, H., & Li, Y.

(2013). XAV939, a tankyrase 1 inhibitior, promotes cell apoptosis in neuroblastoma cell

lines by inhibiting Wnt/β-catenin signaling pathway. Journal of experimental & clinical

cancer research, 32(1), 100.

Tracey, W & Treadway, Judith & Magee, William & Sutt, Jill & McPherson, R & Levy, Carolyn

& Wilder, Donald & Yu, Li & Chen, Yue & Shanker, Ravi & Mutchler, Alison & Smith,

Andrew & Flynn, David & Knight, Delvin. (2004). Cardioprotective effects of ingliforib, a

novel glycogen phosphorylase inhibitor. American journal of physiology. Heart and

circulatory physiology. 286, 1177-1184.

Tsopmo, A., Awah, F. M., & Kuete, V. (2013).Book, Medicinal Plant Research in Africa:

Pharmacology and Chemistry, First Edition. Chapter,12. Lignans and Stilbenes from

African Medicinal Plants. 435–478.

Turnham, R. E. & Scott, J. D. (2016). Protein kinase A catalytic subunit isoform PRKACA;

History, function and physiology. Gene, 577(2), 101–108.

Tyagi, N. ., Sharma, G. N. ., Shrivastava, B. ., Chaudhary, N. ., & Sahu, N. (2017). Cancer: An

Overview. International Journal of Research and Development in Pharmacy & Life

Sciences, 6(5), 2740 – 2747.

Uthandi, A. & Karuppasamy, R. (2012). Hepatoprotective Activity Of Sesame Meal On High Fat

Fed Wistar Rats. Journal of Intercultural Ethnopharmacology. 1, 10, 5455.

Valdman, A., Fang, X., Pang, S.T., Ekman, P. and Egevad, L. (2004) ‗Pim-1 expression in

prostatic intraepithelial neoplasia and human prostate cancer‘, Prostate, 60, 367–371.

Valko, M., Rhodes, C. J., Moncol, J., Izakovic, M., & Mazur, M. (2006). Free radicals, metals

and antioxidants in oxidative stress-induced cancer. ChemicoBiological Interactions,

160(1), 1–40.

Ventola, C. L. (2017). Cancer Immunotherapy, Part 1: Current Strategies and Agents. P & T : a

peer-reviewed journal for formulary management, 42(6), 375–383.

Verhelst, S.H & Bogyo, M. (2005). "Chemical proteomics applied to target identification and

drug discovery". BioTechniques, 38:175−177.

Waaler J, Machon O, Tumova L, Dinh H, Korinek V, Wilson SR, Paulsen JE, Pedersen NM,

Eide TJ, Machonova O. (2012). A novel tankyrase inhibitor decreases canonical Wnt

signaling in colon carcinoma cells and reduces tumor growth in conditional APC mutant

mice. Cancer Res 72, 2822-2832.

Wang, C., Yu, J., Wang, H., Zhang, J., Wu, N. (2014). Lipid peroxidation and altered anti-

oxidant status in breast adenocarcinoma patients. Drug Res. 64, 690–692.

Page 140: Computational Approach Towards Exploring the

424

Wang, H., Lu, B., Castillo, J., Zhang, Y., Yang, Z., McAllister, G., Lindeman, A., Reece-Hoyes,

J., Tallarico, J., Russ, C., Hoffman, G., Xu, W., Schirle, M., & Cong, F. (2016). Tankyrase

Inhibitor Sensitizes Lung Cancer Cells to Endothelial Growth Factor Receptor (EGFR)

Inhibition via Stabilizing Angiomotins and Inhibiting YAP Signaling. The Journal of

biological chemistry, 291(29), 15256–15266.

Wang, J. S., Wang, C. L., Wen, J. F., Wang, Y. J., Hu, Y. B., & Ren, H. Z. (2008). Lithium

inhibits proliferation of human esophageal cancer cell line Eca-109 by inducing a G2/M

cell cycle arrest. World journal of gastroenterology, 14(25), 3982–3989.

Wang, L., Liu, J., Zhang, A., Cheng, P., Zhang, X., Lv, S., Wu, L., Yu, J., di, W., Zha, J., Kong,

X., Qi, H., Zhong, Y., Ding, G. (2012). BVT.2733, a Selective 11β-Hydroxysteroid

Dehydrogenase Type 1 Inhibitor, Attenuates Obesity and Inflammation in Diet-Induced

Obese Mice. PloS one. 7, 40056.

Wang, L., Ma, C., Wipf, P., Liu, H., Su, W., & Xie, X. Q. (2013). TargetHunter: an in silico

target identification tool for predicting therapeutic potential of small organic molecules

based on chemogenomic database. The American Association of Pharmaceutical Scientists

(AAPS) journal, 15(2), 395–406.

Wang, L., Waltenberger, B., Pferschy-Wenzig, E. M., Blunder, M., Liu, X., Malainer, C.,

Blazevic, T., Schwaiger, S., Rollinger, J. M., Heiss, E. H., Schuster, D., Kopp, B., Bauer,

R., Stuppner, H., Dirsch, V. M., & Atanasov, A. G. (2014). Natural product agonists of

peroxisome proliferator-activated receptor gamma (PPARγ): a review. Biochemical

pharmacology, 92(1), 73–89.

Wang, R.; Wang, L.; Li, Y.; Hu, H.; Shen, L.; Shen, X.; Pan, Y.; Ye, T.; Zhang, Y.; Luo, X.

(2014). FGFR1/3 tyrosine kinase fusions define a unique molecular subtype of non-small

cell lung cancer. Clin. Cancer Res. 1, 4107–4114.

Wang, X. & Sun, Z. (2019). Understanding PIM-1 kinase inhibitor interactions with free energy

simulation. Phys. Chem. Chem. Phys. 21, 7544–7558 .

Wang, Y., Ren, F., Li, B., Song, Z., Chen, P., & Ouyang, L. (2019). Ellagic acid exerts antitumor

effects via the PI3K signaling pathway in endometrial cancer. Journal of Cancer, 10(15),

3303–3314.

Wang, Z. P., Che, Y., Zhou, H., Meng, Y. Y., Wu, H. M., Jin, Y. G., Wu, Q. Q., Wang, S. S., &

Yuan, Y. (2020). Corosolic acid attenuates cardiac fibrosis following myocardial infarction

in mice. International journal of molecular medicine, 45(5), 1425–1435.

Wang, Z., Bhattacharya, N., Weaver, M., Petersen, K., Meyer, M., Gapter, L., & Magnuson, N.

S. (2001). Pim-1: a serine/threonine kinase with a role in cell survival, proliferation,

differentiation and tumorigenesis. Journal of veterinary science, 2(3), 167–179.

Page 141: Computational Approach Towards Exploring the

422

Warfel, N. A., & Kraft, A. S. (2015). PIM kinase (and Akt) biology and signaling in tumors.

Pharmacology & therapeutics, 151, 41–49.

Wetherilt H. (1992). Evaluation of urtica species as potential sources of important nutrients.

Developments in food science, 29, 15-25.

World Health Organization (WHO), (2017). "Global report on diabetes".

World Health Organization (WHO), (2019). "Global report on diabetes".

Xavier, C., Cristovao, L.,Ferreira,F., Wilson, P. (2009). Salvia fruticosa, Salvia officinalis, and

rosmarinic acid induce apoptosis and inhibit proliferation of human colorectal cell lines:

the role in MAPK/ERK pathway. Nutrition and Cancer. 61(4):564-71.

Xie, B., Shi, L., Li, J., Zeng, Y., Liu, W., Tang, S., Gan, G. (2019). Oleanolic acid inhibits

RANKL-induced osteoclastogenesis via ER alpha/miR-503/RANK signaling pathway in

RAW264.7 cells. Biomedicine & Pharmacotherapy, 117, 109045 .

Xu, D., Allsop, S.A., Witherspoon, S.M., Snider, J.L., Yeh, J.J., Fiordalisi, J.J., White, C.D

Williams, D., Cox, A.D. and Baines, A.T. (2011) ‗The oncogenic kinase Pim-1 is

modulated by K-Ras signaling and mediates transformed growth and radioresistance in

human pancreatic ductal adenocarcinoma cells‘, Carcinogenesis, 32(4) 488–495.

Xu, H., Zong, H., Ma, C., Ming, X., Shang, M., Li, K., He, X., Du, H., & Cao, L. (2017).

Epidermal growth factor receptor in glioblastoma. Oncology letters, 14(1), 512–516.

Xu, X., Guignard, C., Renaut, J., Hausman, J. F., Gatti, E., Predieri, S., & Guerriero, G. (2019).

Insights into Lignan Composition and Biosynthesis in Stinging Nettle (Urtica dioica L.).

Molecules (Basel, Switzerland), 24(21), 3863.

Xu, Y., & Fisher, G. J. (2012). Receptor type protein tyrosine phosphatases (RPTPs) - roles in

signal transduction and human disease. Journal of cell communication and signaling, 6(3),

125–138.

Yadav, A. K., Kumar, V., Bailey, D. B., & Jang, B. C. (2019). AZD1208, a Pan-Pim Kinase

Inhibitor, Has Anti-Growth Effect on 93T449 Human Liposarcoma Cells via Control of the

Expression and Phosphorylation of Pim-3, mTOR, 4EBP-1, S6, STAT-3 and AMPK.

International journal of molecular sciences, 20(2), 363.

Yang, F., Zheng, T., Gao, Y., Baskota, A., Chen, T., Ran, X., & Tian, H. (2014). Increased

plasma DPP4 activity is an independent predictor of the onset of metabolic syndrome in

Chinese over 4 years: result from the China National Diabetes and Metabolic Disorders

Study. PloS one, 9(3), 92222.

Yang, J., Nie, J., Ma, X., Wei, Y., Peng, Y., & Wei, X. (2019). Targeting PI3K in cancer:

mechanisms and advances in clinical trials. Molecular Cancer, 18(26), 1_28.

Page 142: Computational Approach Towards Exploring the

422

Yang, L., Allred, K. F., Dykes, L., Allred, C. D., & Awika, J. M. (2015). Enhanced action of

apigenin and naringenin combination on estrogen receptor activation in non-malignant

colonocytes: implications on sorghum-derived phytoestrogens. Food & function, 6(3),

749–755.

Yang, Q., Chen, L. S., Neelapu, S. S., & Gandhi, V. (2013). Combination of Pim kinase inhibitor

SGI-1776 and bendamustine in B-cell lymphoma. Clinical lymphoma, myeloma &

leukemia, 13 Suppl 2(0 2), S355–S362.

Yang,W., Chen, X., Li, Y., Guo, S., Wang, Z., and Yu, X. (2019). Advances in Pharmacological

Activities of Terpenoids. Natural Product Communications Vol. 15(3): 1–13.

Yesil-Celiktas, O., Sevimli, C., Bedir, E., & Vardar-Sukan, F. (2010). Inhibitory effects of

rosemary extracts, carnosic acid and rosmarinic acid on the growth of various human

cancer cell lines. Plant foods for human nutrition (Dordrecht, Netherlands), 65(2), 158–

163.

Yokoyama, T., Kosaka, Y., & Mizuguchi, M. (2015). Structural Insight into the Interactions

between Death-Associated Protein Kinase 1 and Natural Flavonoids. Journal of medicinal

chemistry, 58(18), 7400–7408.

Yu, X., Geer, L.Y., Han, L., & Bryant, S.H. (2015). Target enhanced 2D similarity search by

using explicit biological activity annotations and profiles. J Cheminform 7, 55.

Zalckvar, E., Berissi, H., Eisenstein, M., & Kimchi, A. (2009). Phosphorylation of Beclin 1 by

DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL.

Autophagy, 5(5), 720–722.

Zemskova, M., Sahakian, E., Bashkirova, S., & Lilly, M. (2008). The PIM1 kinase is a critical

component of a survival pathway activated by docetaxel and promotes survival of

docetaxel-treated prostate cancer cells. The Journal of biological chemistry, 283(30),

20635–20644.

Zhang, H., Zhai, J., Zhang, L., Li, C., Zhao, Y., Chen, Y., Li, Q., & Hu, X. P. (2016). In Vitro

Inhibition of Glyoxalase І by Flavonoids: New Insights from Crystallographic Analysis.

Current topics in medicinal chemistry, 16(4), 460–466.

Zhang, Z., Zhao, D., Dai, Y., Cheng, M., Geng, M., Shen, J., Ma, Y., Ai, J., & Xiong, B. (2016).

Design, Synthesis and Biological Evaluation of 6-(2,6-Dichloro-3,5-dimethoxyphenyl)-4-

substituted-1H-indazoles as Potent Fibroblast Growth Factor Receptor Inhibitors.

Molecules (Basel, Switzerland), 21(10), 1407.

Zhang, Z.Y. & Lee, S.Y. (2003). PTP1B inhibitors as potential therapeutics in the treatment of

type 2 diabetes and obesity. Expert Opin. Investig. Drug, 12(2):223-233.

Zhao J, Zhao D, Poage GM, Mazumdar A, Zhang Y, Hill JL. (2015). Death-associated protein

kinase 1 promotes growth of p53-mutant cancers. J Clin Invest 125(7): 2707-20.

Page 143: Computational Approach Towards Exploring the

421

Zhao, L., Yuan, X., Wang, J., Feng, Y., Ji, F., Li, Z., & Bian, J. (2019). A review on flavones

targeting serine/threonine protein kinases for potential anticancer drugs. Bioorganic &

medicinal chemistry, 27(5), 677–685.

Zheng, M., Liu, X., Xu, Y., Li, H., Luo, C., & Jiang, H. (2013). Computational methods for drug

design and discovery: focus on China. Trends in pharmacological sciences, 34(10), 549–

559.

Zheng, Y., Ley, SH., and Hu, FB. (2018). "Global aetiology and epidemiology of type 2 diabetes

mellitus and its complications". Nature Reviews Endocrinology, 14:88-89.

Ţiberna, L., Šamec, D., Mocan, A., Nabavi, S. F., Bishayee, A., Farooqi, A. A., Sureda, A., &

Nabavi, S. M. (2017). Oleanolic Acid Alters Multiple Cell Signaling Pathways:

Implication in Cancer Prevention and Therapy. International journal of molecular sciences,

18(3), 643.