Extraction, isolation and characterization of and its analogues from syzygium aromaticum (cloves) and evaluation of their biological activities.

A ―Dissertation submitted in Partial Fulfilment of the Requirements for the Degree of Masters of Science (MSc.) in Chemistry in the Faculty of Science and Agriculture, at the University of Fort Hare‖

By

Khwaza Vuyolwethu (201209227)

Supervisor: Prof. O. O. Oyedeji

Co-supervisor: Prof. B. A. Aderibigbe

2019

i DECLARATION

I, ―Khwaza Vuyolwethu, student number 201209227 declare that this Dissertation entitled,

Extraction, isolation and characterization of oleanolic acid and its analogues from

Syzygium aromaticum (cloves) and evaluation of their biological activities, which I hereby submit to the university of Fort hare in partial fulfilment of the requirements for the Masters of Science (Chemistry) is my own original work, it has never been submitted for any academic award to any other institution of higher learning.‖

……………………………. ……………………………………

Khwaza Vuyolwethu Date

……………………………. ……………………………………

Prof O.O Oyedeji Date

Supervisor

……………………………. ……………………………………

Prof. B. A. Aderibigbe Date

Co-Supervisor

ii

PLAGIARISM DECLARATION

I, Khwaza Vuyolwethu, student number 201209227 hereby declare that I am fully aware of the University of Fort Hare‘s policy on plagiarism and I have taken every precaution to comply with the regulations.

1. I declare that this dissertation is the results of my own original work. Where someone

else‘s work was used (others sources i.e internet, printed sources etc.) due

acknowledgment was given and reference was made according to the department

requirements.

2. I did not use another student‘s work and submit it as my own work.

3. I did not and will not allow anyone to copy my work with the intention of presenting

it as his/her own work.‖

iii

CONFERENCE AND PUBLICATIONS

Conferences

 Vuyolwethu Khwaza, Opeoluwa O. Oyedeji and Blessing A. Aderibigbe (2018).

Isolation and Characterization of oleanolic acid and its analogues from syzygium

aromaticum. Poster presentation at the BIOAFRICA CONVENTION: International

conference Durban ICC South Africa.

Publications

 Vuyolwethu Khwaza, Opeoluwa O. Oyedeji and Blessing A. Aderibigbe (2018).

Antiviral Activities of Oleanolic Acid and Its Analogues. Molecules 2300 (23) 1-14.

 Khwaza V, Opeoluwa O. Oyedeji, Mike O.Ojemaye, Adebola O.Oyedeji and Francis

B.Lewu (2018) ―Assessment of the heavy metal content of wild and cultivated

Pelargonium inquinans: an herbal plant used for the treatment of divers ailments in

South Africa‖. Fresenius Environmental Bulletin 27(6) 3914-165.

iv

ACKNOWLEDGEMENTS

 In Africa they say ―It takes a village to raise a child‖. Nothing in this world is ever

successful without a corporate of many talented people who are willing to work and

submit their talents, experiences, and passion for a common goal.

 Firstly I would like thank God Almighty for making it possible for me to complete

this work in spite of many difficulties experienced along the way.

 I wish to thank my supervisors Prof. O. O. Oyedeji and Prof. B.A. Aderibigbe for

their assistance, guidance and encouragement throughout the study, you both played a

major role in my life. Thank you and may you continue to make tremendous

difference in South Africa and the world at large. It is an honour to have been under

your supervision.

 My Mentor ―from Sasol Dr Du Plessis Esna, thank you for your words of

encouragement you have carried me to where I am today. For that, God bless you and

your family.

 I would like to thank Mr T. Mcako for helping with the FTIR spectroscopy.

 I wish to thank Prof. R. Krause and Dr S. N. Xavier from Rhodes University for their

excellent assistance with NMR, and MS.

 I would like to thank National Research Foundation (NRF), Sasol inzalo foundation

and Medical Research Council for funding.‖

 The Department of chemistry, University of fort hare.

 I would like to send my sincere gratitude to my brother Mtobeli Khwaza ―Madala‖

and my dear sister Tobeka Khwaza for your support, encouragement and for being

there for me throughout.

 A special thanks to Khatywa Ongeziwe. You‘ve been a big brother to me since my

first day in UFH.

v

 I want to thank all my lab mates for lending their expertise when required.

 My family at large thank you for your prayers, it is not my by power that I managed

to finish, but through your prayers. From the bottom of my heart I love you so much.

vi

DEDICATION

I dedicate this work to my Mom (Nozuzile khwaza) for being the positive motivating force in my life. You‘ve struggled against all odds to get me educated. May God Almighty shower you with all the blessings that you deserve, I love you Mazaka.

vii

ABSTRACT

Pathogenic microorganisms have serious impact on people's lives. Every year, millions of people around the world die of bacterial infections. Resistance to common antibacterial drugs has proven to be a challenging problem in control of bacterial infections. In an attempt to develop an effective and affordable treatment for bacterial infections, oleanolic acid isolated from syzygium aromaticum conjugates incorporating other pharmaceutical scaffolds such as chloroquine derivatives, curcumin, and ergocalciferol etc have been developed. Based on the previous successes of testing combination of antimicrobial drugs and pharmaceutical drugs which appeared to be the promising strategy to overcome treatment failure; a series of hybrid compounds containing oleanolic acid and other pharmaceutical scaffolds were synthesized. 4-

Aminoquinoline derivatives were first hybridized with selected organic compounds to form a class of hybrid compounds containing either amide bond or ester bond as a linker between the precursor molecules. Analogues/hybrid compounds can overcome the disadvantages of combination therapy such as drug-drug interaction. The structural effects of this type of conjugation of oleanolic acid and other pharmaceutical scaffolds were characterised by FT-

IR, Mass Spec and NMR spectroscopy. These compounds were studied along with the mono- substituted oleanolic acid analogues and the organic components in order to compare the effects of the substitution on their biological response.‖ All the synthesized analogues were tested against 11 bacterial strains on both Gram-positive and Gram-negative bacteria. The synthesized compounds showed selectivity and higher activity against Enterococcus faecalis

(EF), Klebsiella oxytoca (KO), Escherischia coli (EC), Staphylococcus aureous (SA),

Proteus vulgaris (PV) and Bacillus subtilis (BS) with MIC values; ranging between of 1.25 mg/mL to 0.072 mg/mL.

Key words: Oleanolic acid, Antibacterial, Syzygium aromaticum, Analogues and

Pharmaceutical scaffolds.

viii

LIST OF FIGURES

Figure 2. 1: phenol (parent structure of all phenolic compounds) ...... 13

Figure 2. 2: examples of phenolic compounds ...... 14

Figure 2. 3: biosynthesis of different classes terpenes...... 16

Figure 2. 4: examples of monoterpenes ...... 17

Figure 2. 5: Sesqueterpene compounds ...... 18

Figure 2. 6: examples of diterpene compounds ...... 19

Figure 2. 7: different classes of structures...... 20

Figure 2. 8: Examples of terpene compounds...... 21

Figure 2. 9: Common modern drugs derived from plant sources ...... 22

Figure 2. 10: antimicrobial drugs ...... 24

Figure 2. 11: 3-acetoxy, 28-methyloleanolic acid (2.42), 3-acetoxyoleanolic acid (2.43)...... 27

Figure 2. 12: oleanolic acid derivatives studied for PTP-1B inhibition...... 29

Figure 2. 13: Oleanolic derivatives with anti-HIVactivity ...... 31

Figure 2. 14: derivatives of oleanolic acid with anti-HIV activities...... 32

Figure 2. 15: Previously modified anti-HIV derivatives...... 32

Figure 3. 1: World‘s distribution of syzygium aromaticum (clove) ...... 52

Figure 3. 2: Fresh (A) and dried (B) syzygium aromaticum ...... 53

Figure 3. 3: Clove‘s oil constituents...... 54

Figure 3. 4: TLC plate showing the isolated three compounds from ethylacetate crude extract.

...... 56

Figure 3. 5: formula for calculating Rf value of a compound...... 57

Figure 3. 6: Structure of Eugenol (Eu)...... 58

ix

Figure 3. 7: Structure of oleanolic acid (OA)...... 60

Figure 3. 8: Structure of ...... 62

Figure 4. 1: IR spectrum of N-(3-aminopropyl)-7-chloroquinolin-4-amine...... 74

Figure 4. 2: IR spectrum of N-(2-aminopropyl)-7-chloroquinolin-4-amine (1.2 PDA-Q) ...... 76

Figure 4. 3: IR spectra of N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine...... 77

Figure 4. 4: FT-IR spectrum of 1-(7-Choloroquinolin-4-yl) hydrazine ...... 79

Figure 4. 5: FT-IR spectrum of 2-(7-chloroquinolin-4-ylamino) ethanol...... 80

Figure 4. 6: FT-IR spectrum of 2-(2-(-chloroquinolin-4-ylamino)ethoxy)ethanol (AEE-Q). . 82

Figure 4. 7: FT-IR spectrum of 4-(7-chloroquinolin-4-ylamino)-2-hydroxybenzoic acid ...... 83

Figure 5. 1: structural elucidation of compound VK1 ...... 101

Figure 5. 2: 1IR spectrum of compound VK1...... 102

Figure 5. 3: 1H-NMR spectrum of compound VK1 ...... 103

Figure 5. 4: 13C-NMR spectrum of compound VK1...... 103

Figure 5. 5: LC-MS results of compound VK1 ...... 104

Figure 5. 6: structural elucidation of compound VK2 ...... 104

Figure 5. 7: IR spectra of compound VK2...... 106

Figure 5. 8: 1H NMR Spectrum of compound VK2 ...... 106

Figure 5. 9: 13C NMR spectrum of compound VK2...... 107

Figure 5. 10: LC-MS results of compound VK2...... 107

Figure 5. 11: Structural elucidation of compound VK3 ...... 108

Figure 5. 12: IR spectra of compound VK3...... 109

x

Figure 5. 13: 1H NMR spectrum of compound VK3...... 110

Figure 5. 14: 13C NMR spectrum of compound VK3 ...... 110

Figure 5. 15:LC-MS results of compound VK3 ...... 111

Figure 5. 16: Structural elucidation of compound VK4 ...... 111

Figure 5. 17: IR spectrum of compound VK4...... 113

Figure 5. 18: 1H NMR spectrum of compound VK4 ...... 113

Figure 5. 19: 13C NMR spectrum of compound VK4...... 114

Figure 5. 20: LC-MS results of compound VK4...... 114

Figure 5. 21: Structural elucidation of compound VK5...... 115

Figure 5. 22: IR spectrum of compound VK5...... 116

Figure 5. 23:1H NMR spectrum of compound VK5 ...... 117

Figure 5. 24: 13C NMR spectrum of compound VK5 ...... 117

Figure 5. 25: LC-MS results of compound VK5 ...... 118

Figure 5. 26: Structural elucidation of compound VK6 ...... 118

Figure 5. 27: IR spectrum of compound VK6...... 119

Figure 5. 28: 1H NMR spectrum of compound VK6...... 120

Figure 5. 29: 13C NMR spectrum of compound VK6...... 120

Figure 5. 30: LC-MS results of compound VK6 ...... 121

Figure 5. 31: Structural elucidation of compound VK7 ...... 121

Figure 5. 32: IR spectrum of Compound VK7...... 122

Figure 5. 33: 1H NMR spectrum of compound VK7 ...... 123

Figure 5. 34: 13C NMR spectrum of compound VK7...... 123

Figure 5. 35: LC-MS results of compound VK7 ...... 124

Figure 5. 36: Structural elucidation of compound VK8 ...... 124

Figure 5. 37: IR spectrum of compound VK8...... 125

xi

Figure 5. 38: 1H NMR spectrum of compound VK8 ...... 126

Figure 5. 39: 13C NMR spectrum of compound VK8 ...... 126

Figure 5. 40: LC-MS results of compound VK8 ...... 127

Figure 5. 41: structural elucidation of compound VK9 ...... 127

Figure 5. 42: IR spectrum of compound VK9...... 128

Figure 5. 43: 1H NMR spectrum of compound VK9...... 129

Figure 5. 44: 13C NMR spectrum of compound VK9 ...... 129

Figure 5. 45: LC-MS results of compound VK9 ...... 130

Figure 5. 46: The 96 Well plates (MIC) showing the antibacterial activities against 11 bacterial strains...... 132

Figure 5. 47: Successful hybrid compounds tested for antibacterial activity...... 134

xii

LIST OF SCHEMES

Scheme 4. 1: N-alkylation of 4.7-dichloroquinoline mechanism...... 72

Scheme 4. 2: synthesis of N-(3-aminopropyl)-7-chloroquinolin-4-amine...... 73

Scheme 4. 3: Synthesis of N-(2-aminopropyl)-7-chloroquinolin-4-amine...... 75

Scheme 4. 4: Symthesis of N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine ... 76

Scheme 4. 5: Symthesis of 1-(7-Choloroquinolin-4-yl) hydrazine (Hyd-Q) ...... 78

Scheme 4. 6: Synthesis of 2-(7-chloroquinolin-4-ylamino) ethanol (EA-Q) ...... 79

Scheme 4. 7: Synthesis of 2-(2-(-chloroquinolin-4-ylamino)ethoxy)ethanol (AEE-Q) ...... 81

Scheme 4. 8: Synthesis of 4-(7-chloroquinolin-4-ylamino)-2-hydroxybenzoic acid ...... 82

Scheme 4. 9: Synthesis of compound VK1 ...... 85

Scheme 4. 10: Synthesis of compound VK2 ...... 86

Scheme 4. 11: Synthesis of compound VK3 ...... 88

Scheme 4. 12: Synthesis of compound VK4 ...... 89

Scheme 4. 13: unsuccessful hybrid compounds ...... 99

Scheme 4. 14: Synthesis of compound VK5 ...... 91

Scheme 4. 15: Synthesis of compound VK6 ...... 92

Scheme 4. 16: Synthesis of compound VK7 ...... 94

Scheme 4. 17: Synthesis of compound VK8 ...... 95

Scheme 4. 18: Synthesis of compound VK9 ...... 97

xiii

“LIST OF TABLES

Table 2. 1: Examples of different major classes of phenolic compounds...... 13

Table 2. 2: Some plant species where oleanolic acid was reported, their biological activities, and the plant parts used...... 26

Table 2. 3: Synthesis of Oleanolic acid...... 30

Table 3. 1: Extraction results...... 58

Table 3. 2: 1H and 13C-NMR data of Compound VK121 compared with literature data...... 59

Table 3. 3: 1H and 13C-NMR data of Compound VK122 compared with literature data...... 60

Table 3. 4: 1H and 13C-NMR data of Compound VK123 compared with literature data ...... 62

Table 5. 1: Antibacterial activities of synthesized compounds...... 131"

xiv

LIST OF ABBREVIATION

1.2 PDA: 1.2-diaminopropane

1.2 PDA-Q: N-(2-aminopropyl)-7-chloroquinolin-4-amine

1.3 PDA: 1.3-diaminopropane

1.3 PDA-Q: N-(3-aminopropyl)-7-chloroquinolin-4-amine

AEE: 2(2-Aminoethoxy) ethanol

AEE-Q: 2-(2-(-chloroquinolin-4-ylamino)ethoxy)ethanol

BA:

CC: Column Chromatography

CHCl3: Chloroform cm-1: per centimetre

DMSO: Dimethylsulfoxide

DCC: N.N'-Dicyclohexylcarbodiimide

DCM: Dichloromethane

DET: Diethelenetriamine

DET-Q: N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine.

DMAP: 4-Dimethylaminopyridine

DMAPP: dimethylally pyrophosphate

DMF: Dimethylformide

xv

EA: Ethanolamine

EA-Q: 2-(7-chloroquinolin-4-ylamino) ethanol

EDA: Ethyldiamine

EDDA: 2-(2-(2-aminoethoxy)ethoxy)ethanamine

EtOAc: ethylcetate

Eu: Eugenol

FDA: food drug administration

FGPP: farnesyl geranyl pyrophosphate

FGPPS: farnesyl geranyl pyrophosphate synthase

FT-IR: Fourier-transorm infrared specroscopy

FPP: farnesyl pyrophosphate

FPPS: Farnesyl pyrophosphate synthase

GGPP: Geranylgeranyl pyrophosphate

GGPPS: Geranylgeranyl pyrophosphate synthase

GPPS: Geranyl pyrophosphate synthase

H2SO4: Sulfuric acid

HIV: Human Immunodeficiency Virus

HYD: Hydrazine hydrate

HYD-Q: 1-(7-Choloroquinolin-4-yl) hydrazine

xvi

Hz: Hertz.

IPP: Isopentenyl pyrophosphate

LC-MS: Liquid chromatography mass spectroscopy

LiAlH4: Lithium Aluminium hydride

MeOH: Methanol

MEP: 2C-Methyl-D- erythritol-4-phosphate

MIC: Minimum inhibition concentration

MVA: Mevalonate mmol: milimole

Mp: Melting point

Ms: Mass spectrometry

NME: New molecular entities

NMR: Nuckear magnetic resonance

OA: Oleanolic acid

OH: Hydroxyl ppm: parts per million

PTP: Protein-tyrosine phosphate

Rf: retardation factor

TEA: Triethylamine

xvii

TLC: Thin layer chromatography

SA: South Africa

UA:

WHO: World Health Organisation.

xviii

TABLE OF CONTENTS

DECLARATION ...... ii

PLEGERISM DECLARATION ...... iii

CONFERENCE AND PUBLICATIONS ...... iv

Conferences ...... iv

Publications ...... iv

ACKNOWLEDGEMENTS ...... v

DEDICATION ...... vii

ABSTRACT ...... viii

LIST OF FIGURES ...... ix

LIST OF SCHEMES...... xiii

LIST OF ABBREVIATION ...... xv

CHAPTER ONE ...... 1

INTRODUCTION ...... 1

1.1. Background of the study ...... 1

1.2. Antibacterial drug resistance ...... 3

1.3. Justification of the study ...... 4

1.4. Null hypothesis...... 5

1.5. Aim and Objectives ...... 5

1.5.1. Overall objectives ...... 5

1.6. References ...... 7

xix

CHAPTER TWO ...... 11

LITERATURE REVIEW ...... 11

2.1. Antibacterial resistance...... 11

2.2. Plant natural products ...... 12

2.3. Classes of natural products ...... 13

2.3.1 Phenolic Compounds ...... 13

2.3.2. Terpenes...... 14

2.3.3. Biosynthesis of terpenes ...... 15

2.3.4. Monoterpenes ...... 17

2.3.5. Sesqueterpenes...... 17

2.3.6. Diterpenes ...... 18

2.3.7. Triterpenes ...... 19

2.4. Role of plants in westernised medicine ...... 21

2.5. Natural products as antimicrobial drugs ...... 22

2.6. Combinations of plant natural products with antimicrobial drugs ...... 24

2.6. Oleanolic acid and its analogues ...... 25

2.6.1. Oleanolic acid ...... 25

2.6.2. Analogues of OA ...... 27

2.7. References ...... 34

CHAPTER THREE ...... 52

PHYTOCHEMICAL EXAMINATION OF SYZYGIUM AROMATICUM (L.) MERR.

& PERRY ...... 52

xx

3.1. Introduction ...... 52

3.2. Medicinal use of syzygium aromaticum...... 53

3.3. Chemistry of syzygium aromaticum...... 54

3.4. EXPERIMENTAL ...... 55

3.4.1 Plant Identification ...... 55

3.4.2. Plant preparation and solvent extraction ...... 55

3.4.3. Isolation method ...... 55

3.4.4. 13C and 1 H NMR spectroscopic analysis ...... 57

3.5. Results ...... 58

3.5.1. Physical properties of compound (VK121) ...... 58

3.5.2. Physical properties of compound (VK122) ...... 60

3.5.3. Physical properties of compound VK123 ...... 62

3.6. Discussion ...... 64

3.6.1. Compound (VK121) ...... 64

3.6.2. Compound (VK122) ...... 65

3.6.3. ―Compound (VK123) ...... 65

3.7. References ...... 67

CHAPTER FOUR ...... 71

SYNTHESIS OF OLEANOLIC ACID ANALOGS ...... 71

4.1. Materials ...... 71

4.2. Characterization ...... 71

xxi

4.3. Introduction ...... 72

4.3. Synthesis of 4-aminoquinoline derivatives...... 73

4.4. Synthesis of N-(3-aminopropyl)-7-chloroquinolin-4-amine (1.3 PDA-Q) ...... 73

4.5. Synthesis of N-(2-aminopropyl)-7-chloroquinolin-4-amine (1.2 PDA-Q) ...... 75

4.6. Symthesis of N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine (DET.Q)

...... 76

4.7. Symthesis of 1-(7-Choloroquinolin-4-yl) hydrazine (HYD-Q) ...... 78

4.8. Synthesis of 2-(7-chloroquinolin-4-ylamino) ethanol (EA-Q) ...... 79

4.9. Synthesis of 2-(2-(-chloroquinolin-4-ylamino)ethoxy)ethanol (AEE-Q) ...... 81

4.10. Synthesis of 4-(7-chloroquinolin-4-ylamino)-2-hydroxybenzoic acid ...... 82

4.11. DERIVATIVES OF OLEANOLIC ACID...... 85

4.11.1. Synthesis of hybrid compounds with amide linkers ...... 85

4.11.2 Synthesis of compound VK1 ...... 85

4.11.3. Synthesis of compound VK2 ...... 86

4.11.4. Synthesis of compound VK3 ...... 88

4.11.5. Synthesis of compound VK4 ...... 89

4.12. Synthesis of hybrid compounds with ester linkers...... 91

4.12.1. Synthesis of compound VK5...... 91

4.12.2. Synthesis of compound VK6 ...... 92

4.12.3. Synthesis of compound VK7 ...... 94

4.12.4. Synthesis of compound VK8 ...... 95

4.12.5. Synthesis of compound VK9 ...... 97

xxii

4.12.6. Antibacterial assay ...... 100

4.12.7 Minimum inhibitory concentration (MIC) ...... 100

CHAPTER FIVE ...... 101

RESULTS AND DISCUSION ...... 101

5.1. Structural elucidation of compound VK1 ...... 101

5.2. Structural elucidation of compound VK2 ...... 104

5.3. Structural elucidation of compound VK3 ...... 108

5.4. Structural elucidation of compound VK4 ...... 111

5.5. Structural elucidation of compound VK5 ...... 115

6.6. Structural elucidation of compound VK6 ...... 118

5.7. Structural elucidation of compound VK7 ...... 121

5.8. Structural elucidation of compound VK8 ...... 124

5.9. Structural elucidation of compound VK9 ...... 127

5.10. Minimum inhibitory concentration ...... 130

5.11. Successful hybrid compounds ...... 133

CHAPTER SIX ...... 135

CONCLUSION AND RECOMMENDATIONS ...... 135

6.1. CONCLUSION ...... 135

6.2. RECOMMENDATIONS ...... 135

6.3. References ...... 137

APPENDIX ONE...... 141

xxiii

SPECTRA OF EUGENOL ...... 141

APPENDIX TWO ...... 145

Specta of oleanolic acid...... 145

APPENDIX THREE ...... 149

Specta of Maslinic acid ...... 149

xxiv

CHAPTER ONE

INTRODUCTION

1.1. Background of the study

Plants ―have been the significant source of medicinal treatments throughout history and still continue playing a fundamental role in the primary health of over 80% of the world's developing and underdeveloped countries1–4. Plants produce a large number of organic compounds, the majority of which is believed not to contribute in growth or development of the plant. These compounds are known as natural products or secondary metabolites. The primary metabolites in contrast, such as chlorophyll, simple carbohydrates, nucleotides, or amino acids are found in every plant kingdom and are essential for growth and normal functioning of the plant5. Plants use these natural products in order to defend themselves against herbivores and pathogenic microorganisms6,7.

Natural products have been generally seen as biologically unimportant and have historically received little attention from most plant biologists. Organic chemists, however, have for quite some time been interested in these novel phytochemicals and have examined their chemical properties widely since the 1850s. Investigations of natural products led to the improvement of isolation strategies, spectroscopic approaches to structure elucidation, and synthetic techniques that presently constitute the establishment of current organic chemistry5.

The study of natural products has attracted many researchers to carry out laborious analysis on the plants and to establish a relationship between phytochemicals/chemical composition and therapeutic activities. The isolation of morphine (1.1), as a pure secondary metabolite in

1806 from the extract of Papaver somniferum L. (opium poppy) historically used as a pain

1 reliever was a breaking ground to drug development from plants8. As a result around half of all drugs in medicinal use today are derived from medicinal plants worldwide. For instance, artemisinin (1.2) an antiviral drug isolated from Artimisia annua is used to treat multi-drug- resistant malaria9 and aspirin (1.3), an isolate of willow bark tree Salix alba L., commonly known as acetylsalicylic acid also considered to be one of the most effective analgesic, antipyretic and anti-inflammatory drugs which are commonly used in modern medicine10, other examples of important plant-derived pharmaceutical drugs in use today are codeine

(1.4), atropine (1.5) and quinine (1.6).

CH3 HO H O OH O N H C O O 3 O O H O H H O CH 3 1.3 1.1 HO O 1.2

O

HO N OH N O H O H N O

HO O N 1.6 1.4 1.5 ”

Figure 1. 1: Examples of pharmaceutical drugs isolated from plants.

2

1.2. Antibacterial drug resistance

The ―abusive and intensive use of antibacterial drugs has dramatically increased the occurrence of microbial resistance and has led to an increase of difficult-to-eradicate infections. To solve the problem, combination therapy of two or more antimicrobial drugs has emerged some years ago, in the belief that they can achieve a reversal of microbial resistance with lower quantities of each substance and can also lower the known antimicrobial drugs‘ toxic side-effects. In spite of the many advantages of combination therapy, several reports have proven that it has failed in several patients11. The clinical development of the good combination therapy is expensive and drug-drug interactions may cause additive toxic side effects. Thus modification of the existing drugs through synthesis of analogues could be a noble solution. Combining two or more active pharmacophores into a single molecule

(analogue) can overcome combination therapy while enhancing overall potency12.

In previous years, the testing of combination of antimicrobial drugs and non-antimicrobial compounds (drugs not initially designed for this reason) appears to be another new promising strategy to overcome treatment failures12–15. Therefore, synthesis of a class of analogues/ hybrid compounds containing a natural product oleanolic acid which has antibacterial activities and other pharmaceutical compounds such as 4-aminoquinoline derivatives,

Curcumin, 4-aminosalicylic acid, Ergocalciferol etc can result in a class of potent compounds with effective antibacterial activities.‖

3

1.3. Justification of the study

According ―to the World Health Organisation (WHO), bacterial resistance to various antibiotics is one of the best challenges to human health in this century because of the rising hospitalization and death rates of patients infected with clinically antibiotic resistant around the world16.‖

Plants ―contain a variety of natural products whose chemical structures often unlikely to be formed in laboratories. The pharmacological activities of plants could be based on the antiviral, antibacterial, antipyretic effects of the active compounds in them17,18. The therapeutic effect of plants has encouraged scientists and drug specialists to do a thorough analysis of the plants and to build up a connection between phytochemicals and their therapeutic activities. In spite of the fact that it has been recognized that plant-based drug is one of the surest means of achieving total health care coverage of the world‘s population.

Many natural products have been studied for their effectiveness and safety but their modification to improve their effectiveness is still lacking. An assessment of all food and drug administration (FDA)-approved and new molecular entities (NMEs) shows that natural products and their derivatives are approximately 33 % of all NMEs19. One of the natural products under investigation is oleanolic acid (OA) isolated from syzygium aromaticum, this compound has been reported to have many medical applications. However, to the best information of this researcher, there is no recorded data about a class of analogues/hybrid compounds containing oleanolic acid and other pharmaceutical compounds (such as 4- aminoquinoline derivatives, curcumin, 4-aminosalicilic acid, etc).‖

Recently, plants have become a major significant source of various biofertilizers, herbicides, fungicides20–22etc. These bioactive compounds may be better than synthetic agrochemicals23 and could be significantly safer from health and environmental perspective. Considering this

4 consistently developing demand of phytochemicals and the varieties that exist among them, it is important to develop a new methodology for modifying these compounds to improve their pharmacological activity. Natural products have played a significant role in designing novel drugs due to their massive structural diversity24. The synthetic transformation of phytochemicals to improve their pharmacological activity has attracted many researchers in organic synthesis and medicinal chemistry25.

1.4. Null hypothesis.

Hybridization of oleanolic acid with selected pharmaceutical scaffolds via the C28 carboxylic acid can result in a class of potent compounds with effective antibacterial activities.

1.5.Aim and Objectives

 To isolate and characterize OA from Clove flower buds.

 To prepare a class of hybrid compounds with enhanced biological activities

containing OA and other pharmaceutical scaffolds (e.g. 4-aminoquinoline derivatives,

4-Aminosalicylic acid, curcumin, etc.) via the C28 carboxylic acid to form ester or

amide linkers

1.5.1. Overall objectives

 To ―extract crudes from Syzygium aromaticum flower buds (clove) using different

solvents such as n-Hexane, Dichloromethane(DCM), Ethylacetate (EtOAc), and

Methanol (MeOH).

 To isolate compounds from fraction extracts using Thin Layer Chromatography

(TLC) and Column Chromatography (CC).

5

 To characterize the isolates and semi-synthesize the analogues using spectroscopic

methods, such as Mass spectrometry (MS), Melting point (m.p), Fourier-transform

infrared spectroscopy (FTIR) and Nuclear Magnetic Resonance (NMR).

 To evaluate the biological activities of oleanolic acid and its analogues.‖

6

1.6. References

1. Khazir J, Mir BA, Pilcher L, Riley DL. Role of plants in anticancer drug discovery.

Phytochem Lett. 2014;7(1):173-181. doi:10.1016/j.phytol.2013.11.010

2. Sasidharan S, Chen Y, Saravanan D, Sundram KM, Yoga Latha L. Extraction,

isolation and characterization of bioactive compounds from plants‘ extracts. African J

Tradit Complement Altern Med. 2011;8(1):1-10. doi:10.4314/ajtcam.v8i1.60483

3. Canter PH, Thomas H, Ernst E. Bringing medicinal plants into cultivation :

opportunities and challenges for biotechnology. Trends Biotechnol. 2005;23(4):180-

185. doi:10.1016/j.tibtech.2005.02.002

4. Akhtar MS, Hossain MA, Said SA. Isolation and characterization of antimicrobial

compound from the stem-bark of the traditionally used medicinal plant Adenium

obesum. J Tradit Complement Med. 2017;7(3):296-300.

doi:10.1016/j.jtcme.2016.08.003

5. Croteau R, Kutchan TM, Lewis NG. Secondary Metabolites. Biochem Mol Biol Plants.

2000;7(7):1250-1318. doi:10.1016/j.phytochem.2011.10.011

6. Anulika NP, Ignatius EO, Raymond ES, Osasere O, Hilda A. The Chemistry Of

Natural Product : Plant Secondary Metabolites. Int J Technol Enhanc Emerg Eng Res.

2016;4(8):1-8.

7. Dixon RA. Natural products and plant disease resistance. Nature. 2001;411(June):843-

847.

8. Rishton GM. Natural Products as a Robust Source of New Drugs and Drug Leads: Past

Successes and Present Day Issues. Am J Cardiol. 2008;101(10):43-49.

7

doi:10.1016/j.amjcard.2008.02.007

9. Gurib-Fakim A. Medicinal plants: Traditions of yesterday and drugs of tomorrow. Mol

Aspects Med. 2006;27(1):1-93. doi:10.1016/j.mam.2005.07.008

10. Gilani AH, Atta-ur-Rahman. Trends in ethnopharmacology. J Ethnopharmacol.

2005;100(1-2):43-49. doi:10.1016/j.jep.2005.06.001

11. Taganna JC, Quanico JP, Perono RMG, Amor EC, Rivera WL. Tannin-rich fraction

from Terminalia catappa inhibits quorum sensing (QS) in Chromobacterium

violaceum and the QS-controlled biofilm maturation and LasA staphylolytic activity in

Pseudomonas aeruginosa. J Ethnopharmacol. 2011;134(3):865-871.

doi:10.1016/j.jep.2011.01.028

12. Zacchino SA, Butassi E, Liberto M Di, Raimondi M, Postigo A, Sortino M. Plant

phenolics and terpenoids as adjuvants of antibacterial and antifungal drugs.

Phytomedicine. 2017;37(October):27-48. doi:10.1016/j.phymed.2017.10.018

13. Bush K, Courvalin P, Dantas G, Davies J, Eisenstein B, Huovinen P, George A.

Tackling antibiotic resistance. Nat Rev Microbiol. 2011;9(12):894-896.

doi:10.1038/nrmicro2693

14. Ejim L, Farha MA, Falconer SB, Wildenhain J, Coombes BK,Tyers M. Combinations

of antibiotics and nonantibiotic drugs enhance antimicrobial efficacy. Nat Chem Biol.

2011;7(6):348-350. doi:10.1038/nchembio.559

15. Lehtinen J, Lilius EM. Promethazine renders Escherichia coli susceptible to penicillin

G: real-time measurement of bacterial susceptibility by fluoro-luminometry. Int J

Antimicrob Agents. 2007;30(1):44-51. doi:10.1016/j.ijantimicag.2007.02.019

8

16. Hu H, Han X, Shi X, Wang J, Han L, Chen D, Ji-Zheng H. Temporal changes of

antibiotic-resistance genes and bacterial communities in two contrasting soils treated

with cattle manure. FEMS Microbiol Ecol. 2016;92(2):1-13.

doi:10.1093/femsec/fiv169

17. Debnath B, Somraj W, Das M, Goswami S, Singh MK, Maiti D, Manna K. Role of

plant alkaloids on human health : A review of biological activities. Mater Today Chem.

2018;9:56-72. doi:10.1016/j.mtchem.2018.05.001

18. Eleojo C, Amoo SO, Kudanga T. Opuntia ( Cactaceae ) plant compounds , biological

activities and prospects – A comprehensive review. Food Res Int. 2018;112:328-344.

doi:10.1016/j.foodres.2018.06.047

19. Patridge E, Gareiss P, Kinch MS, Hoyer D. natural products and their derivatives.

Drug Discov Today. 2016;21(2):204-207. doi:10.1016/j.drudis.2015.01.009

20. Cos P, Vlietinck AJ, Vanden D, Maes L. Anti-infective potential of natural products :

How to develop a stronger in vitro ‗ proof-of-concept .‘ J Ethnopharmacol.

2006;106(3):290-302. doi:10.1016/j.jep.2006.04.003

21. Dayan FE, Cantrell CL, Duke SO. Bioorganic & Medicinal Chemistry Natural

products in crop protection. Bioorg Med Chem. 2009;17(12):4022-4034.

doi:10.1016/j.bmc.2009.01.046

22. Koehler AM, Shew HD. Field efficacy and baseline sensitivity of Septoria steviae to

fungicides used for managing Septoria leaf spot of stevia. Crop Prot. 2018;109:95-

101. doi:10.1016/j.cropro.2018.03.006

23. Dossey AT. Natural Product Reports. Nat Prod Rep. 2010;27(12):1725-1944.

9

24. Newman DJ. Natural Products as Leads to Potential Drugs : An Old Process or the

New Hope for Drug Discovery J Med Chem. 2008;51:2589-2599.

25. Dinh T, Moons N, Kim Y, Borggraeve WD, Mashentseva A, Andrei G, Snoeck R.

Synthesis of triterpenoid triazine derivatives from allobetulone and betulonic acid with

biological activities. Bioorg Med Chem. 2014;22(13):3292-3300.

doi:10.1016/j.bmc.2014.04.061.

10

CHAPTER TWO

LITERATURE REVIEW

2.1. Antibacterial resistance

Antimicrobial drugs have for quite some time been instrumental in combating infectious diseases such as cancer, antioxidants, diabetes, etc. ―since the first discovery of antibiotic, penicillin from the plant in 1928 by Alexander Fleming. Antibiotics have proven to be highly effective for the control of infectious diseases, however, the increase in the number of resistant pathogenic microorganisms to antibiotics in the past few years has gradually rendered conventional drugs less effective1. The misuse or abusive of these drugs has also aided in lowering antimicrobial effectiveness. As a result of the increase in resistance, many patients in clinics are being left untreated. According to WHO most people who are being disappointed by orthodox medicine often turn to medicinal plant remedies2.

There are three different types of antibacterial resistance, namely intrinsic, acquired and clinical resistance. Intrinsic resistance is seen when the pathogens are not exposed to the antibiotic. Acquired resistance occurs among the microbes that have been exposed to the antibiotic when managing the infection, and is always due to a genetic mutation. .Clinical resistance occurs when there is a failure in drug, which could be affected by many factors, including immune status of the patients, and the pharmacokinetics of the treatment or the species of pathogens being treated3,4. The emergence of resistance to conventional antimicrobial drugs has become a major problem to public health4. This global concern has led to efforts being directed to finding solutions to the problem of antimicrobial resistance.

One of the strategies to overcome this problem is the identification of new, more effective

11 antimicrobial alternatives. Many studies are thus, being directed towards discovering new antimicrobials, to which no resistance has developed.‖

The ―need for new antibiotic improvement to confront the threat imposed by resistant pathogens has become a major global concern for human health. Most antibiotics in clinical use today are derived from natural products5. To confront the challenge there is a need for the discovery and development of a new class of antibiotics.‖

2.2. Plant natural products

Therapeutic ―plants have shown the great potential of giving powerful or effective drugs for prevention and treatment of infectious diseases with antimicrobial drugs. As a result, a number of researchers are concentrating on plant natural products with the aim to discover novel drugs. Due to limited accessibility or high cost of many antimicrobial drugs in developing countries, most people in these countries rely on traditional medicinal plants6,7.

South Africa (SA) is one of the countries where a number of people rely on medicinal plant especially in rural areas. Hence, there is also a great need to validate how plant isolates and their analogues could lead to the discovery of potent and novel drugs.‖

Plants ―produce a large number of different compounds that seem to have no direct function in the development and growth of the plant, but instead they help the plant to adapt to its environment for the purpose of self-defense and to interact with other organisms in the environment8, these compounds are collectively referred to as natural products or secondary metabolites. Natural products involved in the protection against microorganism such as bacteria, viruses, and fungi, also, some plants use natural products as signals for communication between symbiotic microorganisms and plants, and attraction of seed dispersers and pollinators via colour and scent9,10. Plant natural products are classified into three chemically major groups: phenolics, terpenes, and alkaloids.‖

12

2.3. Classes of natural products

2.3.1 Phenolic Compounds

Phenolic ―compounds are the most widely distributed secondary metabolites, naturally present in the plant kingdom11. The consumption of a diet rich in phenolic compounds has been hypothesized to be important in health promotion and disease prevention in humans and animals12. Phenolic compounds are characterized as aromatic metabolites that have one or more acidic hydroxyl (OH) groups attached to the phenyl ring.‖

OH

2.1

Figure 2. 1: Phenol (parent structure of all phenolic compounds) Polyphenols ―can be divided into different classes depending on their number of carbon atoms and basic chemical structure. Table 2.1 shows the basic chemical structure of different polyphenolic compounds.‖

Table 2. 1: ―Examples of different major classes of phenolic compounds.‖

Class Basic Skeleton Number of C-atoms

Simple phenols, and C6 6 Benzoquinoline

Naphthaquinones C6 – C4 10

Anthraquinones C6 – C2–C6 14

Flavonoids C6 – C3–C6 15

Phenylpropenes (C6 – C5–C6)n n

13

Phenolic ―compounds possess various health benefits including treatment and prevention of many diseases such as cardiovascular disease, cancer, diabetes, and obesity13,14. Other phenolic compounds such as Caffeic acid(2.2) in figure 2 are known to be the strong antiviral15, antioxidant16 and anticancer17 agent. Eugenol (2.3) an isolate of syzygium aromaticum and many other plants also possesses antibacterial and antioxidant activities18,19.‖

O

OH O

HO HO OH 2.3 2.2

Figure 2. 2: Examples of phenolic compounds

2.3.2. Terpenes

The second ―class of natural products or secondary metabolites are the terpenes also known as terpenoids, these compounds are structurally characterized by the presence of at least one or more isoprene units (C5H10 = C5) which are normally joined in a head-to-tail manner. The fusion of isoprene units produces subfamilies of terpenes named as follows: monoterpenes

(C10), sesquiterpenes (C15), diterpenes (C20), sesterpenee (25), triterpenes (C30), and

20 tetraterpenes (C40) and polyterpenes (C5)n, n is the number of isoprene units greater than 8 .‖

14

2.3.3. Biosynthesis of terpenes

Despite the ―massive structural differences between terpenes, they are all derived from isoprene skeleton units. There are two precursors for the synthesis of terpenes which includes isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP). They are synthesized through a different number of rearrangement, cyclization reactions and repeats.

Two different pathways for the formation of both IPP and DMAPP have been reported, 2C- methyl-D-erythritol-4-phosphate (MEP) pathway and the classical mevalonate (MVA) pathway The MVA pathway is present in the chloroplasts of plants. The MVA pathway

(Figure 2.3) includes seven enzymatic reactions to convert acetyl-CoA precursor to IPP and

DMAPP while the MEP pathway converts pyruvate and glyceraldehyde-3-phosphate, to

DMAPP and IPP through eight different enzymatic reactions21,22. The linear prenyl diphosphates: geranylgeranyl pyrophosphate (GGPP), farnesyl pyrophosphate (FPP), farnesyl geranyl pyrophosphate (FGPP) and geranyl pyrophosphate (GPP), are synthesized from the both basic building blocks, DMAPP and IPP where a group of enzymes known as prenyltransferases frequently add the isoprene unit IPP to (DMAPP) or a prenyl diphosphate in consecutive head-to-tail condensations leading to the production of a range of molecules with fixed lengths and stereochemistry. Geranyl pyrophosphate synthase (GPPS) and farnesyl pyrophosphate synthase (FPPS) catalyze the condensation of IPP and DMAPP to produce

GPP (C10) and FPP (C15). Geranylgeranyl pyrophosphate synthase (GGPPS) and farnesyl geranyl pyrophosphate synthase (FGPPS) are responsible for the formation of GGPP (C20) and FGPP (C25). The precursors GPP, FPP, GGPP and FGPP, are rearranged by different terpene synthase enzymes to produce the different classes of terpenoids23,24.‖

15

Mevalonate pathway Non-mevalonate pathway

Pyruvate + D-Glyceraldehyde 3-phosphate

Acetyl-CoA + Acetyl-CoA DXS Acetyl-CoA 1-Deoxy-D-xylulose 5-phosphate (DOXP) thiolase IspC Acetoacetyl-CoA HMG-CoA 2-C-Methyl-D-erythritol 4-phosphate (MEP) synthesis

3-hydroxy-3-methylglutaryl-CoA IspD HMG-CoA reductase 4-diphosphocyltidyl-2C-methyl-D-erythritol

Mevalonate IspE Mevalonate 4-diphosphocyltidyl-2C-methyl-D-erythritol- kinase 2-phosphate Mevalonate 5-phosphate IspF Phosphomevalonate kinase 2C-methyl-D-erythritol-2,4-cyclodiphosphate Mevalonate 5-pyrophosphate IspG Mevalonate ,,,,,,,,,,,,pyrophosphate 1-Hydroxy-2-methyl-2-butenyl 4-diphosphate ,,,,,,,,,,,,,,,decarboxylate

Isopentyl pyrophosphate (IPP) Diethylally pyrophosphate (DMAPP)

IDI

GPPS Monoterpene synthesis Monoterpenes GPP IPP FPPS Sesqueterpene & triterpene synthesis Monoterpenes FPP triterpenes IPP GPPS Diterpenes & Tetrapene synthesis Tetrapernes GGPP Diterpenes 16

Figure 2. 3: Biosynthesis of different classes‖ terpenes25. 2.3.4. Monoterpenes

Monoterpenes (C25) are commonly found in the essential oils and these compounds are essential for the flavouring, cosmetic, and pharmaceutical industries26–28. Figure 2.4 shows examples of monoterpenes include ―camphor (2.5), α-pinene (2.6), 1,8-cineole (2.7), (2.8) and Perillyl alcohol (2.9), and carvone(2.10) and geraniol (2.11).‖

O

O

2.5 2.6 2.7

OH

O OH

2.8 2.9 2.10 2.11

Figure 2. 4: Examples of monoterpenes 2.3.5. Sesquiterpenes

Sesquiterpenes―(C15) are secondary metabolites which are commonly found in various plants with essential oils. Most sesquiterpenes are known to possess a number of biological activities such as germacrene D (2.12) which is known to have anti-inflammatory properties29,30. One of the important sesquiterpene compounds is artemisinin (2.13), an antimalarial drug obtained from Artemisia annua L. (Asteraceae)31,32. This plant has been used to treat malaria and fevers in China for many years33. Other examples of sesquiterpenes

17 shown in figure 2.5 include humulene (2.14), caryophyllene (2.15) lactone (2.16) and δ- cadinene (2.17).”

H

O O O H H O 2.14 2.12 2.13 O

H O

O H H 2.17 2.16 2.15

Figure 2. 5: Sesquiterpene compounds

2.3.6. Diterpenes

Diterpenes (C20) are one of the largest groups of non-volatile terpenes formed from the fusion of four isoprene units. Recent studies showed that oral administration of diterpene trans-retinoic acid (2.18) could be used as alternative treatment for skin flap necrosis in diabetic patients undergoing flap surgery34. Diterpenes like taxol (2.19) possesses some biological activities such as anticancer to treat breast, prostate and ovarian cancers. Taxol, and forskolin(2.20) the isolates of Pacific Yew (Taxus brevifolia) and coleus plant

(Plectranthus scutellarioides) respectively are common major diterpenes obtained from nature28,35. Diterpenes are divided into different major classes such as acyclic, bicyclic, tricyclic, tetracyclic and the macrocyclic diterpenes. Jatrophone (2.21), an isolate of elliptica

18

J. and Jatrapus (J.) gossypifolia is an example of macrocyclic diterpene and was reported to inhibit tumor cells36. ―

O OH

COOH O

OH 2.18 H 2.20 O

OH O O O O O

O O O O O H O O O O O 2.21 O O O O 2.19

Figure 2. 6: Examples of diterpene compounds

2.3.7. Triterpenes

Triterpenes (C30) are formed from the fusion of 6 isoprene units and widely distributed in nature including plants, animals, microorganisms, and humans. Triterpenes are one of the largest classes of secondary metabolites synthesized through cyclization of squalene37.

Structurally, terpenes have four 6-membered rings (A, B, C, D) and E being 5-membered or

6-membered ring and are divided into 6 different common subgroups as shown in figure 6: (2.22), ursane (2.23), friedelane (2.24), gammacerane(2.25) and lupine (2.26), hopane (2.27)38.

19

E E E C D C D C D

A B A B A B

2.22 2.23 2.24

E E E C D C D C D A B A B A B

2.25 2.26 2.27

Figure 2. 7: Different classes of triterpenes structures.

Triterpenes possessed various therapeutic properties, such as antiviral39,40, antimicrobial41,42, antioxidant43–45, and anti-inflammatory46,47. Examples of triterpenes include ursolic acid UA

(2.28), oleanolic acid OA (2.29), lupeol (2.30) and betulinic acid BA (2.31) as shown in

Figure 2.8. OA and BA acid are triterpenes commonly known to have an inhibitory effect on the human immunodeficiency virus (HIV) pathogen48,49.

20

OH OH

O O

HO HO 2.29 2.28

OH

O

HO 2.30 HO 2.31

Figure 2. 8: Examples of terpene compounds.

2.4. Role of plants in westernized medicine

Most common modern drugs were derived from plant sources, including quinine (2.32), aspirin (2.33), morphine (2.34), and atropine (2.35), and codeine (2.36). According to

Phillipson (2001), worldwide about 50% of clinically used top 20 drugs, are derived from plants. Until now, novel modern drugs are still being derived from plants sources. The number of phytomedicines entering the market increases worldwide50.

21

HO HO N O OH O N O O H N 2.32 O 2.33 HO 2.34

O

N OH

O O H 2.36 H N 2.35 O HO

Figure 2. 9: Common modern drugs derived from plant sources

2.5. Natural products as antimicrobial drugs

Bacterial ―resistance to antibiotics is rapidly emerging around the world, endangering the effectiveness of antibiotics and now causing a serious problem to human health51–53. The extensive use and abusive/misuse of antibiotics in developing countries has increased the occurrence of antibiotic resistance to make it a worldwide‖issue54,55. To confront this challenge an urgent development of novel drugs and antimicrobial strategies are needed.

There are different potential next-generation therapeutic strategies such as antibacterial peptides produced by microorganisms56, metal nanoparticles57, bacteriophages58 and plant natural products, such as terpenes, phenols, and alkaloids obtained in many plants52.

In order to find more effective drugs for microbial therapy, research has been directed to the medicinal plant. These plants have been proven over centuries to be successfully inhibiting infectious diseases among our ancestors, they provide a promising source of novel

22 antimicrobial drugs59,60. In many developing countries such as South Africa, therapeutic plants play an important role in human health and ―about 80% of people in developing countries use medicinal plants because of its cultural acceptability and affordability61,62. It has been reported that plant-derived antimicrobial drugs are rarely associated with side-effects, and furthermore have the ability to treat many types of diseases63. The use of the plant as antimicrobial treatment was first reported in Europe with the discovery of ―Iceman‖ the body of a human being that was preserved by bracket fungus64.‖

Plants ―have been known for centuries as a source of medicine to treat‖ microbial diseases and the use of the plant as a medicine is well documented. Rios and Recio 2005 also noted an increased number of research articles being published on the use of plant-derived antimicrobial drugs65. The main focus of many studies is on the determination of antimicrobial efficacy of the plants and also included the antimicrobial screening of plant extracts66,67 and essential oils68–70. Other studies have gone as far to test the isolated antimicrobial active compounds from plants against different pathogens, many of these studies are listed in article reviews by Cowan (1999)71 and Van Vuuren (2008)72and (2017)73.

Most plant-derived natural products such as pentacyclic triterpenes have a great potential to inhibit microbial pathogens as they are active against various bacterial species of both Gram- negative and Gram-positive and they directly target the cell envelope52. Triterpenes are one of the largest and structurally diverse groups of secondary metabolites with approximately 200 different compounds74. These compounds are commonly found in medicinal plants and food in free form/bound to . Triterpenes and their analogues have been studied for their antimicrobial75,76, anti-inflammatory77, antivirals78,79, and antioxidant characteristics80.

Chemical compounds obtained from plants have always been investigated by the pharmaceutical companies as agents of biological properties, or as models for the synthesis of novel drugs such as antimicrobials81.‖

23

2.6. Combinations of plant natural products with antimicrobial drugs

Most natural products were found to be synergistic enhancers for antimicrobial drugs, even if the plant natural compounds are non-antimicrobial82. Other studies have investigated the effects of combining plant natural products (e.i plant extracts and essential oils) and antimicrobial drugs such as cefuroxime (2.37), tobramycin (2.38), tetracycline (2.39), amphotericin B (2.40), and nystatin (2.41).

HO NH2

NH HO 2 H H S O NH2 N HO O H2N N O OH O O OH O 2.38 O NH2 2.37 H2N OH

OH OH O OH O OH O O OH OH H NH2 HO O OH OH OH OH O OH

OH O H H HO H N O O 2.39 2.40

HO OH OH NH OH 2 O OH H HO O OH OH OH OH O OH

O H O O 2.41

HO OH

NH2

Figure 2. 10: Antimicrobial drugs.

24

These were tested on a range of resistant microbes, such as methicillin-resistant

Pseudomonas aeruginosa and Staphylococcus aureus (MRSA)83–85, a synergistic interaction has been noted in most cases. Some of these studies have focused on antibiotic combinations with plant extracts of Rosmarinus officinalis L.86, Melaleuca alternifolia87,88, Origanum vulgare, Thymus vulgaris, Mentha piperita89and Syzygium aromaticum90. The synergistic effect is shown by a reduced minimum inhibition concentration (MIC) for the antimicrobial and the reduced MIC indicates an improved antimicrobial effect, which can finally render an ineffective antimicrobial drug to be effective again and this has resulted to define other plant extracts as resistance modifying ―agents91. Van Vuuren et al. (2011) and Adwan et al. (2010) argued that the enhanced effect made by plant extracts on antimicrobial drugs has been ignored and it needs further investigation92,93.‖

2.6. Oleanolic acid and its analogues

2.6.1. Oleanolic acid

Oleanolic acid (3b-Hydroxy-olea-12-en-28-oic acid) (2.29), is a bioactive natural compound found in various plants and foods, it belongs to the triterpene family of‖ compounds. The role of OA in plants is often associated with the prevention of pathogens and water loss76.

Oleanolic acid possesses various interesting biological activities, such as anti-inflammatory, analgesic47,94, antibacterial75,95, antiviral78,96, antitumor97 anti-cancer98–101, anti- oxidation102,103, and cardioprotective activities101. Chen et al. 2014 also reported that OA offers extraordinary protection against chronic and acute injury, and could be used in oral administration for human liver disorders104,105. Oleanolic acid has been isolated from more than 1600 various plant species76,104,106–108, and is moderately water soluble and non-toxic47.

Some medicinal plants containing oleanolic acid as their active constituent are demonstrated in table 2.2 below.

25

Table 2. 2: Some plant species where oleanolic acid was reported, their biological activities, and the plant parts used.

Plant Species (Family) Biological Activity Plant Parts Used References

Aceriphyllum rossii Stems, leaves, Cytotoxic Anticomplement activity 109,110 (Saxifragaceae) roots

Astilbe chinensis Cytotoxic rhizomes 111 (Saxifragaceae)

Baccharis uncinella Antileishmanial Aerial 112,113 (Asteraceae)

Fabiana imbricata R. et Antiviral, antitumor, and Leaves and 114–116 P. (Solanaceae) antihyperlipidemic flowers

Fructus Ligustri Lucidi Anti-hepatitis Leaves 96,117 (FLL)

Gentiana lutea Dried root and Antimicrobial 118 (Gentianaceae) rhizome

Anti-inflammatory, antioxidative, Ligustrum lucidum Ait antiprotozoal, antimutagenic, and Fruits and leaves 119 (Oleaceae) anticancer

L. camara Anti-inflammatory, antioxidative, Leaves and 119 (Verbenaceae) antiprotozoal flowers

Oleaeuropaea L. Anticancer, antimicrobial, anti-diabetic Fruits and leaves 76,102,120 (Oleaceae)

Phyllanthus amarus Anti-diabetes Leaves or aerial 121 (Phyllanthaceae)

Punica granatum L. Antioxidant activity Fruit 101,102 (Punicaceae)

Rosa laevigata Anti-inflammatory Leaves 122 (Rosaceae)

Rosmarinus officinalis Hepatoprotective, Anti-inflammatory, Leaves, flowers, 102 L. (Lamiaceae) gastroprotective, antiulcer stems, branches.

Siphonodon celastrineus Anti-inflammatory Root bark, stem 123,124 (Celastraceae)

26

Syzygium aromaticum Antinociceptive, Anti-inflammatory, Flower buds and 47,96,114,125 (Myrtaceae) antioxidant and antihypertensive, leaves

Viburnum chingii Antimicrobial Leaves 126 (Asteraceae)

Viscum album Anti-tumor, anti-inflammatory and Leaves and stems 119,127 (Santalaceae) analgesic

2.6.2. Analogues of OA

OA contains three(3) active sites (i.e., the hydroxyl, alkene, and carboxylic acid), which can be decorated in order to improve its biological effects122,128,129. Most derivatives of OA have been synthesized or tested for many biological activities130. OA is a good precursor molecule for synthesis due to its various biological activities, availability, and low production cost131.

Nkeh-Chungag et al 2015 reported ―the acetylation and methylation of OA initially isolated from Syzygium aromaticum (clove), which resulted to the formation two compounds (3- acetoxyoleanolic acid and 3-acetoxy, 28-methyloleanolic acid) as shown in figure (2.11).

Both compounds exhibited better membrane-stabilizing properties and anti-inflammatory properties when compared to OA77.

Compound R OR H 2.42 CH O O 3 H O 2.43 H H

Figure 2. 11: 3-acetoxy, 28-methyloleanolic acid (2.42), 3-acetoxyoleanolic acid (2.43).

27

Modification of a compound OA has resulted in compounds with various biological activities such as ant-diabetic, antiviral antimicrobial activities, etc. Yolanda et al. 2014 synthesized derivatives of OA (3 ethers and 4 esters on C3 in ring A, 3 esters from C-28 and corresponding primary alcohol) by reduction of carboxylic acid (COOH) with LiAlH4.

Cinnamoyl ester (2.49) and ethyl ether (2.53) (Figure 2.12) were found to be the most PTP-

1B inhibitors. The in vitro inhibitory effect of compound (2.53) was significant and it substantially lowered blood glucose levels in vivo experiments when compared to OA.

Compound (2.53) exhibited better inhibitory activity and selectivity over the protein-tyrosine phosphatase 1B (PTP-1B) with advanced interaction with site B, in accordance with docking studies121,132.

Compound R1 R2

2.44 COCH3 H

2.45 H CH3

2.46 O H

2.47 H CH2CH3

2.48 CO(CH2)2CH3 H

2.49 O H

2.50 H H2C

28

2.51 H H

2.52 CH3 H

OR2 H 2.53 CH2CH3 H

O H 2.54 (CH2)2CH3 H

R1O H

Figure 2. 12: Oleanolic acid derivatives studied for PTP-1B inhibition.

The modification of oleanolic acid also resulted in potent antibacterial agents. Hichri et al. explored the effect of introducing an acyl substituent at the hydroxyl C-3 in ring A of OA. A sequence of diverse triterpenic acid esters was prepared from oleanolic acid using suitable cyclic anhydrides, acid chlorides and N,N-dimethyl-4-aminopyridine (DMAP) as a catalyst

(table 2.3)118.

Oleanolic acid and its acylated analogues were screened for their antimicrobial activity against five fungal plant pathogens, two Gram-positive and two Gram-negative bacteria.

Compound (2.55) with sulfur and chlorine atom(s): ((3b)-3-((thiophene-2-carbonyl)oxy)- olean-12-en-28-oic acid, was found to be an effective antibacterial agent and the most active antifungal compound. It exhibited good activity against A. niger, P. italicum, P. digitatum, A. flavus, and T. harzianum.

H H OH OH O H O X /DMAP H HO H RO H

29

Table 2. 3: Synthesis of Oleanolic acid.

Compound R X Yield(%)2

O 2.55 RCl 98 S

Cl O 2.56 RCl 94 Ph

Cl O 2.57 RCl 91

O Cl 2.58 RCl 95 Cl

2.59 RCl 94

O O 2.60 O O 92 HOOC

O O 2.61 82 O COOH O

O O 2.62 O 91 COOH O

O 2.63 85 HOOC

30

Synthesis of oleanolic acid, as well as other closely-related triterpenes, such as betulinic acid and dihydrobetulinic acid, has led to anti-HIV agents48,79,133 . Zhu et al. synthesized derivatives of OA, These authors modified the C12-C13 double bond of OA yielding compound 2.64, which was 3-fold more active than OA. Esterification of 2.64 with anhydrides resulted in compounds 2.65–2.67, which were 5-fold more active than OA with

2.67 showing remarkable activity49.

Compound R

2.64 H

H

2.65 HOOC H COOH O

2.66 HOOC

RO O

2.67 HOOC O

Figure 2. 13: Oleanolic derivatives with anti-HIV activity

31

Compound 2.64 was further modified by converting the C28-carboxyl group to an aminomethyl group, resulting in compounds 2.68 and 2.69, which were greater than 10-fold more active when compared to OA (Figure 2.13).

Compound R1 R2

H R2 2.68 HO O O HN O O OH

R1O

2.69 HO O O O O HN OH

Figure 2. 14: Derivatives of oleanolic acid with anti-HIV activities.

Yu et al. in their structure-activity relationship study of effective anti-HIV agents synthesized and evaluated new triterpene derivatives in vitro for antiviral activity. OA analogue compound (2.70) was inactive, while OA derivative (2.71) exhibited an EC50 value of 0.32

μM, indicating that OA is a promising anti-HIV inhibitor49,134. These compounds are potential therapeutics that would benefit from further studies in vivo (Figure 2.15).

Compound R OH 2.70 O O O OH H O RO O O O H 2.71 OH

Figure 2. 15: Previously modified anti-HIV triterpene derivatives.

32

Kashiwada et al. prepared several 3-O-acyl-ursolic acids and evaluated their anti-HIV

135 activity. The most potent compound indicated an EC50 value of 0.31 µM and a TI of 155.5 .

In another report by Kashiwada et al., OA derivatives inhibited HIV-1 replication in acutely infected H9 cells. OA-triterpenes isolated from the leaves of S. claviflorum exhibited potent anti-HIV activity, which further revealed the potential of OA derivatives for the treatment of

HIV136,137.‖

33

2.7. References

1. Morehead MS, Scarbrough C. Emergence of Global Antibiotic Resistance. Prim Care

Clin Off Pract. 2018;45(3):467-484.

2. WHO (World Health Organization)., Antimicrobial resistance – Fact Sheet No. 194.

Retrieved from http://www.who.int/medicentre/factsheets/fs194/en/index.html

(Accessed 04.11.12). 2002.

3. Ambrose SJ, Harmer CJ, Hall RM. Evolution and typing of IncC plasmids contributing

to antibiotic resistance in Gram-negative bacteria. Plasmid. 2018.

doi:10.1016/j.plasmid.2018.08.001

4. Macgowan A. Antibiotic resistance. Medicine (Baltimore). 2017;45(10):622-628.

doi:10.1016/j.mpmed.2017.07.006

5. Townsley L, Shank EA. Natural-Product Antibiotics: Cues for Modulating Bacterial

Biofilm Formation. Trends Microbiol. 2017;25(12):1016-1026.

doi:10.1016/j.tim.2017.06.003

6. Tang S, Prodhan ZH, Biswas SK, Le C, Sekaran SD. Antimicrobial peptides from di ff

erent plant sources : Isolation , characterisation , and purifi cation. Phytochemistry.

2018;154:94-105. doi:10.1016/j.phytochem.2018.07.002

7. Arulmozhi P, Vijayakumar S, Kumar T. Phytochemical analysis and antimicrobial

activity of some medicinal plants against selected pathogenic microorganisms. Microb

Pthogenes. 2018;123(July):219-226. doi:10.1016/j.micpath.2018.07.009

8. Kennedy DO, Wightman EL. Herbal Extracts and Phytochemicals : Plant Secondary

Metabolites and the Enhancement of Human Brain Function 1. Am Soc Nutr.

34

2011;2:32-50. doi:10.3945/an.110.000117.32

9. Salam AM, Quave CL. ScienceDirect Opportunities for plant natural products in

infection control. Curr Opin Microbiol. 2018;45:189-194.

doi:10.1016/j.mib.2018.08.004

10. Yang L, Wen K, Ruan X, Y Zhao Y, Wei F, Wang Q. Response of Plant Secondary

Metabolites to Environmental Factors. Molecules. 2018;23(762):1-26.

doi:10.3390/molecules23040762

11. Cheynier V, Comte G, Davies KM, Lattanzio V, Martens S. Plant phenolics: Recent

advances on their biosynthesis, genetics, andecophysiology. Plant Physiol Biochem.

2013;72:1-20. doi:10.1016/j.plaphy.2013.05.009

12. Ramful D, Bahorun T, Bourdon E, Tarnus E, Aruoma OI. Bioactive phenolics and

antioxidant propensity of flavedo extracts of Mauritian citrus fruits: Potential

prophylactic ingredients for functional foods application. Toxicology. 2010;278(1):75-

87. doi:10.1016/j.tox.2010.01.012

13. Chacko SM, Thambi PT, Kuttan R, Nishigaki I. Beneficial effects of green tea : A

literature review. Chin Med. 2010;13(5):1-9.

14. Zhang Y, Gan R, Li S, Zhou Y, Li A, Xu D. Antioxidant Phytochemicals for the

Prevention and Treatment of Chronic Diseases. Molecules. 2015;20:21138-21156.

doi:10.3390/molecules201219753

15. Wu Z, Yu Z, Cui Z, Peng L, Li H, Zhang C, Shen H, Yi P, Fu B. In vitro antiviral ef fi

cacy of caffeic acid against canine distemper virus. Microb Pathog. 2017;110:240-244.

doi:10.1016/j.micpath.2017.07.006

35

16. Shiozawa R, Inoue Y, Murata I, Kanamoto I. Effect of antioxidant activity of caffeic

acid with cyclodextrins using ground mixture method. Asian J Pharm Sci.

2018;13(1):24-33. doi:10.1016/j.ajps.2017.08.006

17. Singh P, Singh A, Pandita D, Lather V. Future Journal of Pharmaceutical Sciences

Synthesis and evaluation of a series of caffeic acid derivatives as anticancer agents.

Futur J Pharm Sci. 2017;xxx:1-7. doi:10.1016/j.fjps.2017.11.002

18. Bonilla J, Poloni T, Lourenço R V, Sobral PJA. Antioxidant potential of eugenol and

ginger essential oils with gelatin / chitosan fi lms ☆. Food Biosci. 2018;23:107-114.

doi:10.1016/j.fbio.2018.03.007

19. Modjinou T, Versace D, Abbad-andaloussi S, Langlois V, Renard E. Antibacterial and

antioxidant photoinitiated epoxy co-networks of resorcinol and eugenol derivatives.

Mater Today Commun. 2017;12:19-28. doi:10.1016/j.mtcomm.2017.03.005

20. Gutiérrez-del-río I, Fernández J, Lombó F. International Journal of Antimicrobial

Agents Plant nutraceuticals as antimicrobial agents in food preservation : terpenoids ,

polyphenols and thiols. 2018;52:309-315. doi:10.1016/j.ijantimicag.2018.04.024

21. Lange BM, Rujan T, Martin W, Croteau R. Isoprenoid biosynthesis : The evolution of

two ancient and distinct pathways across genomes. Proc Natl Acad Sci USA.

2000;97(24):13172-13177.

22. Eisenreich W, Schwarz M, Zenk MH, Bacherl A, Cartayrade A, Arigoni D. The

deoxyxylulose phosphate pathway of terpenoid biosynthesis in plants and

microorganisms. Chem Biol. 1998;5(9):R221–R233.

23. Wand G, Tang W, Bidigare RR. Terpenoids As Therapeutic Drugs and Pharmaceutical

Agents. Drug Discov Ther Med. 2005:197–227. doi:10.1007/978-1-59259-976-9

36

24. Tholl D. Terpene synthases and the regulation , diversity and biological roles of

terpene metabolism. Curr Opin Plant Biol. 2006;9(3):297-304.

doi:10.1016/j.pbi.2006.03.014

25. Abdallah II, Quax WJ. A Glimpse into the Biosynthesis of Terpenoids. Int Conf Nat

Resour Life Sci. 2016;2017:81-98. doi:10.18502/kls.v3i5.981

26. Jovanovi KK, Sokovi M. Chemical composition , antimicrobial , and cytotoxic

properties of five Lamiaceae essential oils Nevenka Gligorijevi c sa Radulovi c. Ind

Crop Prod jo. 2014;61:225-232. doi:10.1016/j.indcrop.2014.07.011

27. Sueishi Y, Nii R. Monoterpene ‘ s multiple free radical scavenging capacity as

compared with the radioprotective agent cysteamine and amifostine. Bioorg Med

Chem Lett. 2018;28(18):3031-3033. doi:10.1016/j.bmcl.2018.08.003

28. Dhanarasu S. Chromatography and Its Applications.; 2012.

29. Adio AM. Germacrenes A – E and related compounds : thermal , photochemical and

acid induced transannular cyclizations. Tetrahedron. 2009;65(8):1533-1552.

doi:10.1016/j.tet.2008.11.050

30. Chen K, Hsieh P, Hwang T, Chang F. Anti-inflammatory Furanogermacrane

sesquiterpenes from Neolitsea parvigemma. Nat Prod Res. 2005;19(3):283-286.

doi:10.1080/14786410410001714669

31. Suresh N, Haldar K. Mechanisms of artemisinin resistance in Plasmodium falciparum

malaria. Curr Opin Pharmacol. 2018;42:46-54. doi:10.1016/j.coph.2018.06.003

32. Darwish RM, Aburjai T, Al-khalil S, Mahafzah A. Screening of antibiotic resistant

inhibitors from local plant materials against two different strains of Staphylococcus

37

aureus. 2002;79:359-364.

33. Çitoğlu GS, Acıkara ÖB. Column Chromatography for Terpenoids and Flavonoids.

Intech open. 2012;2:13-64. doi:10.5772/32009

34. Weng Z, Wang C, Zhang C, Xu J, Chai Y, Jia Y. All-trans retinoic acid improves the

viability of ischemic skin flaps in diabetic rat models. Diabetes Res Clin Pract.

2018;142:385-392. doi:10.1016/j.diabres.2018.06.019

35. Cretton S, Saraux N, Monteillier A, Righia D, Marcourta L, Genta-Jouveb G,

Wolfendera J, Cuendeta M, Christen P. Anti-inflammatory and antiproliferative

diterpenoids from Plectranthus scutellarioides. Phytochemistry. 2018;154:39-46.

doi:10.1016/j.phytochem.2018.06.012

36. Devappa RK, Makkar HP, Klaus B. Jatropha Diterpenes : a Review. J Am Oil Chem

Soc. 2011;88(3):301-322. doi:10.1007/s11746-010-1720-9

37. Ruzicka L. The isoprene rule and the biogenesis of terpenic compounds. Experientia.

1953;9(10):357-367. doi:10.1007/BF02167631

38. Xiao S, Tian Z, Wang Y, Si L, Zhang L, Zhou D. Recent progress in the antiviral

activity and mechanism study of pentacyclic triterpenoids and their derivatives. Med

Res Rev. 2018;38:1-26. doi:10.1002/med.21484

39. Tsuji M, Sriwilaijaroen N, Inoue H, Miki K, Kinoshita K. Bioorganic & Medicinal

Chemistry Synthesis and anti-influenza virus evaluation of triterpene-sialic acid

conjugates. Bioorg Med Chem. 2018;26(1):17-24. doi:10.1016/j.bmc.2017.09.038

40. Khwaza V, Oyedeji OO, Aderibigbe BA. Antiviral Activities of Oleanolic Acid and Its

Analogues. molecules. 2018;2300(23):1-14. doi:10.3390/molecules23092300

38

41. Laure A, Pagning N, Tamokou J, Lateef M, Tapondjou LA, Kuiate J, Ngnokam D.

New triterpene and new fl avone glucoside from Rhynchospora corymbosa (

Cyperaceae ) with their antimicrobial , tyrosinase and butyrylcholinesterase inhibitory

activities. Phytochem Lett. 2016;16:121-128. doi:10.1016/j.phytol.2016.03.011

42. Khumalo GP, Sadgrove NJ, Vuuren SF Van, Wyk B Van. South African Journal of

Botany Antimicrobial lupenol triterpenes and a polyphenol from Elaeodendron

transvaalense , a popular southern African medicinal bark. South African J Bot.

2018;xx:4-7. doi:10.1016/j.sajb.2018.07.020

43. Qiu J, Wang X, Song C. Fitoterapia Neuroprotective and antioxidant lanostanoid

triterpenes from the fruiting bodies of Ganoderma atrum. Fitoterapia. 2016;109:75-79.

doi:10.1016/j.fitote.2015.12.008

44. Xu J, Wang X, Su G, Yue J, Sun Y, Cao J. The antioxidant and anti-hepatic fi brosis

activities of acorns ( Quercus liaotungensis ) and their natural galloyl triterpenes. J

Funct Foods. 2018;46(February):567-578. doi:10.1016/j.jff.2018.05.031

45. Smina TP, Mathew J, Janardhanan KK, Devasagayam TPA. Antioxidant activity and

toxicity profile of total triterpenes isolated from Ganoderma lucidum ( Fr .) P . Karst

occurring in. Environ Toxicol Pharmacol. 2011;32(3):438-446.

doi:10.1016/j.etap.2011.08.011

46. Rajamani K, Balasubramanian T. Bioassay-guided isolation of triterpene from brown

alga Padina boergesenii possess anti-in fl ammatory and anti-angiogenic potential with

kinetic inhibition of β -carotene linoleate system. LWT - Food Sci Technol.

2018;93:549-555. doi:10.1016/j.lwt.2018.04.010‖

47. Rali S, Oyedeji OO, Aremu OO, Oyedeji AO, Nkeh-Chungag BN. Semisynthesis of

39

derivatives of oleanolic acid from Syzygium aromaticum and their antinociceptive and

anti-inflammatory properties. Mediators Inflamm. 2016;2016:1-9.

doi:10.1155/2016/8401843

48. Zhao H, Holmes SS, Baker GA, Challa S, Bose HS Song Z. Ionic derivatives of

betulinic acid as novel HIV-1 protease inhibitors. J Enzyme Inhib Med Chem.

2012;27(5):715-721. doi:10.3109/14756366.2011.611134

49. Zhu Y, Shen J, Wang H, Mark L, Lee K. Synthesis and Anti-HIV Activity of

Oleanolic Acid Derivatives. Bioorg Med Chem Lett. 2001;11:3115-3118.

50. Cordell GA, Colvard MD. Some thoughts on the future of ethnopharmacology. J

Ethnopharmacol. 2005;100:4-14. doi:10.1088/1751-8113/40/12/S19

51. Wright GD. Antibiotic resistance in the environment : a link to the clinic ? Curr Opin

Microbiol. 2010;13(5):589-594. doi:10.1016/j.mib.2010.08.005

52. Kurek A, Nadkowska P, Pliszka S, Wolska KI. Modulation of antibiotic resistance in

bacterial pathogens by oleanolic acid and ursolic acid. Phytomedicine. 2012;19(6):515-

519. doi:10.1016/j.phymed.2011.12.009

53. Ventola CL. The Antibiotic Resistance Crisis Part 1 : Causes and Threats. Pharm Ther.

2015;40(4):277-283.

54. Taganna JC, Quanico JP, Perono RMG, Amor EC, Rivera WL. Tannin-rich fraction

from Terminalia catappa inhibits quorum sensing (QS) in Chromobacterium violaceum

and the QS-controlled biofilm maturation and LasA staphylolytic activity in

Pseudomonas aeruginosa. J Ethnopharmacol. 2011;134(3):865-871.

doi:10.1016/j.jep.2011.01.028

40

55. Morgan DJ, Okeke IN, Laxminarayan R, Perencevich EN, Weisenberg S. Non-

prescription antimicrobial use worldwide : a systematic review. Lancet Infect Dis.

2011;11(9):692-701. doi:10.1016/S1473-3099(11)70054-8

56. Giuliani A, Pirri G, Nicoletto SF. Antimicrobial Peptides : An Overview of a

Promising Class of Therapeutics. Vol 2.; 2007. doi:10.2478/s11535-007-0010-5

57. Kurek A, Grudniak AM, Anna KD, Krystyna IW. New Antibacterial Therapeutics and

Strategies. Polish J Microbiol. 2011;60(1):3-12.

58. Chibani-chennoufi S, Sidoti J, Bruttin A, Kutter E, Sarker S, Bru H. In Vitro and In

Vivo Bacteriolytic Activities of Escherichia coli Phages : Implications for Phage

Therapy. Antimicrob Agents Chemother. 2004;48(7):2558-2569.

doi:10.1128/AAC.48.7.2558

59. Rabe T, Van Staden J. Antibacterial activity of South African plants used for

medicinal purposes. J Ethnopharmacol. 1997;56:81-87.

60. Cowan MM. Plant Products as Antimicrobial Agents. 1999;12(4):564-582.

61. Wyk AS Van, Prinsloo G. Medicinal plant harvesting, sustainability and cultivation in

South Africa. Biol Conserv. 2018;227:335-342. doi:S0006320718310863

62. Maroyi A. Traditional use of medicinal plants in south-central Zimbabwe: Review and

perspectives. J Ethnobiol Ethnomed. 2013;9(31):18. doi:10.1186/1746-4269-9-31

63. Chanda S, Rakholiya K. Combination therapy : Synergism between natural plant

extracts and antibiotics against infectious diseases. Sci against Microb Pathog

Commun Curr Res Technol Adv. 2011:520-529.

64. Capasso L. 5300 years ago, the Ice Man used natural laxatives and antibiotics. Lancet.

41

1998;352:1864. doi:10.1016/S0140-6736(05)79939-6

65. Ríos JL, Recio MC. Medicinal plants and antimicrobial activity. J Ethnopharmacol.

2005;100(1-2):80-84. doi:10.1016/j.jep.2005.04.025

66. Yuan W, Lee HW, Yuk HG. Antimicrobial efficacy of Cinnamomum javanicum plant

extract against Listeria monocytogenes and its application potential with smoked

salmon. Int J Food Microbiol. 2017;260:42-50. doi:10.1016/j.ijfoodmicro.2017.08.015

67. Santoro D, Ahrens K, Vesny R, Navarro C, Gatto H, Marsella R. Evaluation of the in

vitro effect of Boldo and Meadowsweet plant extracts on the expression of

antimicrobial peptides and inflammatory markers in canine keratinocytes. Res Vet Sci.

2017;115:255-262. doi:10.1016/j.rvsc.2017.05.021

68. Zheljazkov VD, Kacaniova M, Dincheva I, Radoukovae T, Semerdjievaf IB, Astatkieg

T, Schlegel V. Essential oil composition, antioxidant and antimicrobial activity of the

galbuli of six juniper species. Ind Crops Prod. 2018;124:449-458.

doi:10.1016/j.indcrop.2018.08.013

69. Ait Babahmad R, Aghraz A, Boutafda A, Papazoglouc EG, Tarantilisd PA, Kanakisd

C, Hafidia M, Ouhdouche Y, Outzourhitf A, Ouhammou A. Chemical composition of

essential oil of Jatropha curcas L. leaves and its antioxidant and antimicrobial

activities. Ind Crops Prod. 2018;121:405-410. doi:10.1016/j.indcrop.2018.05.030

70. Rodrigues FFG, Colares AV, Nonato C de FA, Galvão-Rodrigues FF, Mota ML,

Braga MFBM, Da Costa JGM. In vitro antimicrobial activity of the essential oil from

Vanillosmopsis arborea Barker (Asteraceae) and its major constituent, α-bisabolol.

Microb Pathog. 2018;125:144-149. doi:10.1016/j.micpath.2018.09.024

71. M, Murphy, Cowan. Plant products as antimicrobial agents. Clin Microbiol Rev.

42

1999;12(4):564–582. doi:0893-

72. Van Vuuren SF. Antimicrobial activity of South African medicinal plants. J

Ethnopharmacol. 2008;119(3):462-472. doi:10.1016/j.jep.2008.05.038

73. Vuuren S Van, Holl D. Antimicrobial natural product research : A review from a South

African perspective for the years 2009 – 2016. J Ethnopharmacol. 2017;208:236-252.

doi:10.1016/j.jep.2017.07.011

74. Hill RA, Connolly JD. Triterpenoids Robert. Nat Prod Reports. 2013;29:780-818.

doi:10.1039/c3np70032a

75. Hichri F, Ben H, Cheriaa J, Jegham S, Mighri Z. Antibacterial activities of a few

prepared derivatives of oleanolic acid and of other natural triterpenic compounds.

Comptes Rendus Chim. 2003;6:473-483. doi:10.1016/S1631-0748(03)00066-3

76. Jesus JA, Lago JHG, Laurenti MD, Yamamoto ES, Passero LFD. Antimicrobial

activity of oleanolic and ursolic acids: An update. Evidence-based Complement Altern

Med. 2015;2015:14. doi:10.1155/2015/620472

77. Nkeh-chungag BN, Oyedeji OO, Oyedeji AO, Ndebia EJ. Anti-Inflammatory and

Membrane-Stabilizing Properties of Two Semisynthetic Derivatives of Oleanolic Acid.

Inflammation. 2015;38(1):61-69. doi:10.1007/s10753-014-0007-y

78. Oleanolic acid and ursolic acid: Novel hepatitis C virus antivirals that inhibit NS5B

activity. Antiviral Res. 2013;98(1):44-53. doi:10.1016/J.ANTIVIRAL.2013.02.003

79. Aiken C, Chen CH. Betulinic acid derivatives as HIV-1 antivirals. TRENDS Mol Med.

2005;11(1):32-36. doi:10.1016/j.molmed.2004.11.001

80. Jeong H, Rae H, Kyong H, Eun Bee Jung, Hyun Bong Park, Ki Sung Kang, Ki Hyun

43

Kim . Bioactivity-guided isolation of antioxidant triterpenoids from Betula platyphylla

var . japonica bark. Bioorg Chem. 2016;66:97-101. doi:10.1016/j.bioorg.2016.04.001

81. Silva N, Fernandes Júnior A. Biological properties of medicinal plants: a review of

their antimicrobial activity. J Venom Anim Toxins Incl Trop Dis. 2010;16(3):402-413.

doi:10.1590/S1678-91992010000300006

82. Aiyegoro OA, Okoh AI. Use of bioactive plant products in combination with standard

antibiotics : Implications in antimicrobial chemotherapy. J Med Plants Res.

2009;3(13):1147-1152.

83. Sato M, Tanaka H, Yamaguchi R, Kato K, Etoh H. Synergistic effects of mupirocin

and an isoflavanone isolated from Erythrina variegata on growth and recovery of

methicillin-resistant Staphylococcus aureus. Int J Antimicrob Agents. 2004;24:43-48.

doi:10.1016/j.ijantimicag.2004.03.020

84. Braga LC, Leite AAM, Xavier KGS, Takahashi JA, Bemquerer MP, Nascimento

AMA. Synergic interaction between pomegranate extract and antibiotics against

Staphylococcus aureus. Can J Microbiol. 2005;51:541-547. doi:10.1139/W05-022

85. Rosato A, Vitali C, Laurentis N De, Armenise D, Antonietta M. Antibacterial effect of

some essential oils administered alone or in combination with Norfloxacin.

Phytomedicine. 2007;14:727-732. doi:10.1016/j.phymed.2007.01.005

86. Jarrar N, Abu-hijleh A, Adwan K. Antibacterial activity of Rosmarinus officinalis L .

alone and in combination with cefuroxime against methicillin-resistant Staphylococcus

aureus. Asian Pac J Trop Med. 2010;3(2):121-123. doi:10.1016/S1995-

7645(10)60049-1

87. Shin S. Anti-Aspergillus Activities of Plant Essential Oils and Their Combination

44

Effects with Ketoconazole or Amphotericin B. Arch Pharm Res. 2003;26(5):389-393.

88. Arrigo MD, Ginestra G, Mandalari G, Furneri PM, Bisignano G. Phytomedicine

Synergism and postantibiotic effect of tobramycin and Melaleuca alternifolia ( tea tree

) oil against Staphylococcus aureus and Escherichia coli. Phytomedicine. 2010;17:317-

322. doi:10.1016/j.phymed.2009.07.008

89. Vuuren SF Van, Suliman S, Viljoen AM. The antimicrobial activity of four

commercial essential oils in combination with conventional antimicrobials. Lett Appl

Microbiol. 2009;48:440-446. doi:10.1111/j.1472-765X.2008.02548.x

90. Elaine J, Betoni C, Mantovani RP, Barbosa LN, Di Stasi LC, Junior AF. Synergism

between plant extract and antimicrobial drugs used on Staphylococcus aureus diseases.

Mem Inst Oswaldo Cruz. 2006;101(June):387-390.

91. Sibanda T, Okoh AI. The challenges of overcoming antibiotic resistance : Plant

extracts as potential sources of antimicrobial and resistance modifying agents. african

J Biotechnol. 2007;6(25):2886-2896.

92. Adwan G, Abu-Shanab B, Adwan K. Antibacterial activities of some plant extracts

alone and in combination with different antimicrobials against multidrug-resistant

Pseudomonas aeruginosa strains. Asian Pac J Trop Med. 2010;3(4):266-269.

doi:10.1016/S1995-7645(10)60064-8

93. Van Vuuren S, Viljoen A. Plant-based antimicrobial studies methods and approaches

to study the interaction between natural products. Planta Med. 2011;77(11):1168-

1182. doi:10.1055/s-0030-1250736

94. Bednarczyk-Cwynar B, Zaprutko L, Marciniak J, Lewandowski G, Szulc M,

Kaminska E, Wachowiak N, Mikolajczak PL. The analgesic and anti-inflammatory

45

effect of new oleanolic acid acyloxyimino derivative. Eur J Pharm Sci.

2012;47(3):549-555. doi:10.1016/j.ejps.2012.07.017

95. Fontanay S, Grare M, Mayer J, Finance C, Duval RE. Ursolic , oleanolic and betulinic

acids : Antibacterial spectra and selectivity indexes ଝ. J Ethnopharmacol.

2008;120(2):272-276. doi:10.1016/j.jep.2008.09.001

96. Kong L, Li S, Liao Q. Oleanolic acid and ursolic acid : Novel hepatitis C virus

antivirals that inhibit NS5B activity. Antiviral Res. 2013;98(1):44-53.

doi:10.1016/j.antiviral.2013.02.003

97. Oprean C, Mioc M, Csányi E. Improvement of ursolic and oleanolic acids ‘ antitumor

activity by complexation with hydrophilic cyclodextrins. Biomed Pharmacother.

2016;83:1095-1104. doi:10.1016/j.biopha.2016.08.030

98. Chouaïb K, Romdhane A, Delemasure S. Regiospecific synthesis , anti-inflammatory

and anticancer evaluation of novel 3 , 5-disubstituted isoxazoles from the natural

maslinic and oleanolic acids. Ind Crop Prod. 2016;85:287-299.

doi:10.1016/j.indcrop.2016.03.024

99. Guo Y, Han B, Luo K, Ren Z, Cai L, Sun L. NOX2-ROS-HIF-1α signaling is critical

for the inhibitory effect of oleanolic acid on rectal cancer cell proliferation. Biomed

Pharmacother. 2017;85:733-739. doi:10.1016/j.biopha.2016.11.091

100. Shanmugam MK, Dai X, Kumar AP, Tan BKH, Sethi G, Bishayee A. Oleanolic acid

and its synthetic derivatives for the prevention and therapy of cancer: Preclinical and

clinical evidence. Cancer Lett. 2014;346(2):206-216. doi:10.1016/j.canlet.2014.01.016

101. Pattnaik B, Nayak L, Sistla R, Mallavadhani V. Bioorganic Chemistry Synthesis of

ring-C modified oleanolic acid derivatives and their cytotoxic evaluation. Bioorg

46

Chem. 2016;68:152-158. doi:10.1016/j.bioorg.2016.08.001

102. Fu Q, Zhang L, Cheng N, Jia M, Zhang Y. Extraction optimization of oleanolic and

ursolic acids from pomegranate (Punica granatum L.) flowers. Food Bioprod Process.

2014;92(3):321-327. doi:10.1016/j.fbp.2012.12.006

103. Bernatoniene J, Cizauskaite U, Ivanauskas L, Jakstas V, Kalveniene Z, Kopustinskiene

DM. Novel approaches to optimize extraction processes of ursolic, oleanolic and

rosmarinic acids from Rosmarinus officinalis leaves. Ind Crops Prod. 2016;84:72-79.

doi:10.1016/j.indcrop.2016.01.031

104. Chen P, Zeng H, Wang Y, et al. Low Dose of Oleanolic Acid Protects against

Lithocholic Acid – Induced Cholestasis in Mice : Potential Involvement of Nuclear

Factor-E2-Related Factor 2-Mediated Upregulation of Multidrug Resistance-

Associated Proteins s. Am Soc Pharmacol Exp Ther. 2014;42:844-852.

105. Aisha AFA, Abu-salah KM, Salman A. Syzygium aromaticum extracts as good source

of betulinic acid and potential anti-breast cancer. Brazilian J Pharmacogn.

2012;22(2):335-343.

106. Sheng H, Sun H. Synthesis, biology and clinical significance of pentacyclic

triterpenes: a multi-target approach to prevention and treatment of metabolic and

vascular diseases. Nat Prod Reports. 2011;28:543–593.

107. J. Liu. Oleanolic acid and ursolic acid: research perspectives. J Ethnopharmacol.

2005;100:92-94. doi:10.1016/j.ijsrc.2016.03.004

108. Fukushima EO, Seki H, Ohyama K, Ono E, Umemoto N, Mizutani M, Kazuki Saito,

Muranaka T. CYP716A Subfamily Members are Multifunctional Oxidases in

Triterpenoid Biosynthesis. Plant Cell Physiol. 2011;52(12):2050-2061.

47

doi:10.1093/pcp/pcr146

109. Van LTK, Hung TM, Thuong PT, Ngoc TM, Kim JC, Jang H. Oleanane-Type

Triterpenoids from Aceriphyllum rossii and Their Cytotoxic Activity. J Nat Prod.

2009;72:1419-1423.

110. Min BS. Anticomplementary Activity of Oleanane-type Triterpenes from the Roots of

Aceriphyllum rossii. Arch Pharm Res. 2012;35(6):1003-1008. doi:10.1007/s12272-

012-0607-8

111. Franch M, Sun H, Ye Y, Pan Y. Cytotoxic oleanane triterpenoids from the rhizomes of

Astilbe chinensis (Maxim.) Franch. et Savat. J Ethnobiol. 2004;90:261-265.

doi:10.1016/j.jep.2003.10.003

112. Passero LFD, Bonfim-melo A, Corbett CEP. Anti-leishmanial effects of purified

compounds from aerial parts of Baccharis uncinella C . DC . ( Asteraceae ). Parasitol

Res. 2011;108:529-536. doi:10.1007/s00436-010-2091-8

113. Yamamoto ES, Campos BLS, Laurenti MD. Treatment with triterpenic fraction

purified from Baccharis uncinella leaves inhibits Leishmania ( Leishmania )

amazonensis spreading and improves Th1 immune response in infected mice.

Parasitol Res. 2014;113:333-339. doi:10.1007/s00436-013-3659-x

114. Sánchez M, Theoduloz C, Schmeda-hirschmann G, Razmilic I, Yáñez T, Rodríguez

JA. Gastroprotective and ulcer-healing activity of oleanolic acid derivatives : In vitro –

in vivo relationships. Life Sci. 2006;79:1349-1356. doi:10.1016/j.lfs.2006.03.044

115. Rodr JA, Astudillo L, Schmeda-hirschmann G. Oleanolic acid promotes healing of

acetic acid-induced chronic gastric lesions in rats. Pharmacol Res. 2003;48:291-294.

doi:10.1016/S1043-6618(03)00155-5

48

116. Quispe C, Viveros-valdez E, Schmeda-hirschmann G. Phenolic Constituents of the

Chilean Herbal Tea Fabiana imbricata R . et P . Plant Foods Hum Nutr.

2012;(67):242-246. doi:10.1007/s11130-012-0302-4

117. Hang YZ, Ai WL, Eung PL, Chun-fu WU, Ao XY. Effects of Fructus Ligustri Lucidi

Extract on Bone Turnover and Calcium Balance in Ovariectomized Rats. Biol Pharm

Bull. 2006;29(2):291-296.

118. Chouab K, Hichri F, Nguir A. Semi-synthesis of new antimicrobial esters from the

natural oleanolic and maslinic acids. Food Chem. 2015;183:8-17.

doi:10.1016/j.foodchem.2015.03.018

119. Banik RM, Pandey DK. Optimizing conditions for oleanolic acid extraction from

Lantana camara roots using response surface methodology. Ind Crops Prod.

2008;27(3):241-248. doi:10.1016/j.indcrop.2007.09.004

120. Strehle A, Thomas C, Sato H. Anti-hyperglycemic activity of a TGR5 agonist isolated

from Olea europaea. Biochem Biophys Res Commun 362. 2007;362:793-798.

doi:10.1016/j.bbrc.2007.06.130

121. Ali H, Houghton PJ, Soumyanath A. ␣ -Amylase inhibitory activity of some

Malaysian plants used to treat diabetes ; with particular reference to Phyllanthus

amarus. J Ethnopharmacol. 2006;107(3):449-455. doi:10.1016/j.jep.2006.04.004

122. Yan M, Zhu Y, Zhang HJ. Anti-inflammatory secondary metabolites from the leaves

of Rosa laevigata. Bioorganic Med Chem. 2013;21(11):3290-3297.

doi:10.1016/j.bmc.2013.03.018

123. Niampoka C, Suttisri R, Bavovada R, Takayama H, Aimi N. Potentially cytotoxic

triterpenoids from the root bark of Siphonodon celastrineus Griff. Arch Pharm Res.

49

2005;28(5):546-549. doi:10.1007/BF02977756

124. Kaweetripob W, Mahidol C, Prawat H, Ruchirawat S. Lupane, friedelane, oleanane,

and ursane triterpenes from the stem of Siphonodon celastrineus Griff. Phytochemistry.

2013;96:404-417. doi:10.1016/j.phytochem.2013.09.027

125. Somova LO, Nadar A, Rammanan P, Shode FO. Cardiovascular , antihyperlipidemic

and antioxidant effects of oleanolic and ursolic acids in experimental. Phytomedicine.

2003;10(2-3):115-121.

126. Chen X-Q, Li Y, He J. Triterpenoids and diterpenoids from Viburnum chingii. Chem

Pharm Bull. 2011;59(4):496-498. doi:10.1248/cpb.59.496

127. Jäger S, Winkler K. Solubility Studies of Oleanolic Acid and Betulinic Acid in

Aqueous Solutions and Plant Extracts of Viscum album L . Planta Med. 2007;73:157-

162. doi:10.1055/s-2007-967106

128. Liby KT, Sporn MB. Synthetic Oleanane Triterpenoids : Multifunctional Drugs with a

Broad Range of Applications for. Pharmacol Rev. 2012;64(4):1-32.

doi:10.1124/pr.111.004846

129. Activity A, Suh N, Wang Y. A Novel Synthetic Oleanane Triterpenoid , 2-Cyano-3 ,

12-dioxoolean-1 , 9-dien-28-oic Acid , with Potent Differentiating , Antiproliferative ,

and. cancer Res. 1999;59:336-341.

130. Chen J, Liu J, Zhang L, Wu G, Hua W. Pentacyclic triterpenes . Part 3 : Synthesis and

biological evaluation of oleanolic acid derivatives as novel inhibitors of glycogen

phosphorylase. Bioorg Med Chem Lett. 2006;16:2915-2919.

doi:10.1016/j.bmcl.2006.03.009

50

131. Pollier J, Goossens A. Phytochemistry Oleanolic acid. Phytochemistry. 2012;77:10-15.

doi:10.1016/j.phytochem.2011.12.022

132. Yolanda M, Paoli P, Jos J. Synthesis of oleanolic acid derivatives : In vitro , in vivo

and in silico studies for PTP-1B inhibition *. Eur J Med Chem. 2014;87:316-327.

doi:10.1016/j.ejmech.2014.09.036

133. Yogeeswari P, Sriram D. Betulinic Acid and Its Derivatives: A Review on their

Biological Properties. Curr Med Chem. 2005;12(6):657-666.

doi:10.2174/0929867053202214

134. Yu D, Sakurai Y, Chen C-H. Anti-AIDS Agents 69. Moronic Acid and Other

Triterpene Derivatives as Novel Potent Anti-HIV Agents. J Med Chem.

2006;49(18):5462-5469.

135. Li J, Guo W, Yang Q. Effects of ursolic acid and oleanolic acid on human colon

carcinoma cell line HCT15. World J Gastroenterol. 2002;8(3):493-495.

136. Sultana N, Ata A. Oleanolic acid and related derivatives as medicinally important

compounds. J Enzyme Inhib Med Chem. 2008;23(6):739-756.

doi:10.1080/14756360701633187

137. Chinsembu KC, Hedimbi M. A Survey of Plants with Anti-HIV Active Compounds

and their Modes of Action. Med J Zambia. 2009;36(4):178-186.

51

CHAPTER THREE

PHYTOCHEMICAL EXAMINATION OF SYZYGIUM AROMATICUM (L.) MERR. &

PERRY

3.1. Introduction

Syzygium ―aromaticum (L.) Merr. & Perry belongs to the Myrtaceae family and is an evergreen tree that grows up to a height of 8-12 meters and is native in Maluku Island of

Indonesia1–4. This plant is also cultivated in many tropical countries such as India, Malaysia,

Zanzibar, and Sri Lanka5. The flower buds of this tree are commonly known as clove and used as a spice. Figure 3.1 shows the native and exotic areas of the plant Syzygium aromaticum.‖

Figure 3. 1: World‘s distribution of Syzygium aromaticum (clove)6

52

3.2. Medicinal use of Syzygium aromaticum.

The ―plant is used as a folk medicine in China and in many western countries against various diseases, such as oral diseases and dental complaints7. The plant is used to treat nausea and vomiting, diarrhea, cough, dyspepsia, stomach distension, flatulence, and gastrointestinal spasm8. According to Pandey and Singh 2011, the cloves are also used for stomachic, tonicardiac, and stimulant effects9. Essential oil obtained from this plant add aroma to the food and serves as a dietary antioxidant which should prevent some diseases caused by free radicals10. The plant is also known to have antibacterial activities and is used in many dental creams, mouth washes, tooth pastes, and throat sprays to cleanse bacteria4,11,12. Many studies revealed it to possess a variety of other biological activities such as anticancer5,13, antioxidant4,14, antifungal15,16, and antidiabetic17. Cloves contain a number of phytochemical compounds such as tannins, sesquiterpenes, Sterols, Flavonoids and triterpenoids11,18.‖

A B

Figure 3. 2: Fresh (A) and dried (B) Syzygium aromaticum6

53

3.3. Chemistry of Syzygium aromaticum.

Syzygium ―aromaticum is one of the major sources of phenolic compounds, like flavonoids, hydroxybenzoic acids, hydroxyphenyl propens and hydroxycinnamic acids, as well as terpenoids19,20. Eugenol constitutes 72 to 90% of the volatile oil in clove and is responsible for the aroma of cloves20. Other common constituents of clove essential oil include β- caryophyllene(3.1), eugenyl acetate(3.2), pinene(3.3), methyl salicylate(3.4), vanillin(3.5) and α-humulene (3.6)10,21.‖

O

O

3.1 3.2 3.3

H O

O

O O OH OH 3.4 3.5 3.6

Figure 3. 3: Clove‘s oil constituents.

54

3.4. EXPERIMENTAL

3.4.1 Plant Identification

―The dried flower buds of Syzygium Aromaticum (L.) were purchased from the spice market in Durban, South Africa. The taxonomic identification of S. aromaticum (L.) was done in the

School of Biological and Conservation Sciences, University of KwaZulu-Natal, Westville

Campus. The voucher specimen number OO4 was deposited at the University Herbarium.‖

3.4.2. Plant preparation and solvent extraction

―The plant material was grounded to a powder form using mortar and pestle. A dried powdered sample (500g) was taken into a 5000 ml Erlenmeyer flask, and sequentially extracted with 2000 ml organic solvents of different increasing polarity namely, n-hexane, ethyl acetate, dichloromethane and methanol. The mixture was placed on a rotating shaker for a period of about two weeks twice per solvent. The supernatant was filtered using filter paper

(Whatman size: 32.0 cm), the filtrate was concentrated on a rotary evaporator at reduced pressure, the concentrated extracts were collected in a pre-weighed beaker and allowed to air dry for complete evaporation of the extracting solvents then air dried in the fume hood. After drying, the weight and the percentage yield of each extract was determined.‖

3.4.3. Isolation method

―The ethyl acetate crude extract (15.535 g) was taken for further purification since the previous studies showed that it contains the mixtures of Maslinic and Oleanolic Acid22. The

Ethyl Acetate crude extract was purified by column chromatography using silica gel (Merck, silica gel 60 F254: 0.063-0.200 mm). The column was eluted with series of solvent systems: n- hexane: ethyl acetate (9:1, 8:2, 7:3 and 6:4) respectively. All fractions were collected and visualized on a Thin Layer Chromatography (TLC) plate with Anisaldehyde/sulphuric acid

55 spray reagent for spot development. All the fractions with similar behaviour on TLC plate, such as fraction 6-33, fraction 40-120, and fraction 123- 173 were combined together and further purified. Combination and purification of these fractions afforded a pale yellow oil pure compound labelled VK121, and two white powder pure compounds labelled VK122 and

VK123.‖

Solvent from

Origin

R vk121 vk122 vk123 CcC3 TLC plate showing the isolated compounds

Figure 3. 4: TLC plate showing the isolated three compounds from ethylacetate crude extract.

―The TLC plate reveals a mixture of compounds, which exhibited different colours when reacting to the anisaldehyde/H2SO4 spraying reagent. The classes of the isolated compounds include the terpenoids, which are bluish or purple in colour when spotted on a TLC plate

(VK122 and VK123)23. The isolated compounds labelled (VK121, VK122 and VK123) were characterized for structural elucidation using Mass Spectrometry, melting point (m.p),

Fourier transform infrared spectroscopy (FT-IR) and nuclear magnetic resonance spectrometry (NMR). The retardation factor (Rf value) of all the isolated compounds was calculated using the formula in figure 3.5.‖

56

Distance travelled by compound

Rf value = Distance travelled by Solvent (Solvent Front)

Figure 3. 5: Formula for calculating Rf value of a compound.

3.4.4. 13C and 1 H NMR spectroscopic analysis

1H and 13C NMR experiments were performed at 400.0 MHz for hydrogen and 13C, using

CDCl3 as solvents. Solutions were prepared with between 15–20 mg of pure compounds in

0.5 ml of CDCl3.

57

3.5. Results

The table below shows the physical state and the % yields of the different crude extracts from cloves of syzygium aromaticum.

Table 3. 1: Extraction results.

Extracting solvents Physical state % Yield

Hexane Dark brown oil 15.6

Ethyl acetate Light orange powder 4.7

DCM light brown oil 1.52

Methanol Dark brown oil 9.7

3.5.1. Physical properties of compound (VK121)

Physical state: Pale yellow liquid.

% Yield: 0.42

Molecular weight:

IR vmax: OH stretch at 3452.37, C-H stretch at 2975 & 2938, ar C=C at 1511.36, C-O at 1033.59.

O 7 2 HO 1 3

6 4 9

5 8 10 Eu

Figure 3. 6: Structure of Eugenol (Eu).

58

Table 3. 2: 1H and 13C-NMR data of Compound VK121 compared with literature data24.

Position δCLit δHLit δCEu δHEu

1 144.3 ---- 142.7 ----

2 114.7 ---- 151.3 ---

3 121.7 6.74(s) 114.5 6.40(s)

4 138.1 ------131.1 ----

5 115.6 6.76 (d, 8.6 Hz) 122.8 6.45(d, 8 Hz)

6 146.9 6.92 (d, 8.6 Hz) 116.8 6.50(d, 8 Hz)

7 111.7 3.86 (s) 56.2 3.73(s)

8 56.3 3.38 (d, 6.6 Hz) 48.4 3.22 (d, 8 Hz)

9 40.3 5.96 (m) 136.5 6.30

10 132.1 5.12(d, 19.7 Hz) 117.2 4.96(d, 8)

5.19(d, 19.7 Hz) 4.93(d, 8)

OH 4.89(s) 5.0(s)

59

3.5.2. Physical properties of compound (VK122)

Physical state: White powdery

% Yield: 0.17

Melting point: 240 oC

Molecular weight: 456.3598

IR vmax: OH stretch at 3443, C-H at 2941 & 2870, C=O at 1693, ar C=C at 1468, C-O at

1031-998.

30 29

19 20 21 12 18 17 22 11 OH 25 26 13 1 9 14 16 28 2 O 10 8 15 3 5 4 HO 7 27 OA 6 23 24

Figure 3. 7: Structure of oleanolic acid (OA).

Table 3. 3: 1H and 13C-NMR data of Compound VK122 compared with literature data25.

25 25 Position δCOA δHOA δC δH

1 33.64 0.99, 1.62 38.37 0.98, 1.62

2 27.48 1.56, 1.60 27.15 1.56, 1.60

3 79.03 3.22(dd, 12, 4 Hz) 79.01 3.20(dd, 12.0, 4.3 Hz)

60

4 41.25 ------38.74 -----

5 55.24 0.81 55.18 0.75

6 22.97 1.36, 1.55 18.27 1.38, 1.54

7 35.3 1.29, 1.36 32.59 1.29, 1.44

8 39.9 ---- 39.23 -----

9 49.8 1.54 47.6 1.54

10 37.03 ------37.05 -----

11 25.88 0.91:1.79 23.37 0.91, 1.88

12 123.07 5.35(t, 4 Hz) 122.61 5.28 (t 3.6 Hz)

13 143.22 ----- 143.26 -----

14 41.25 ----- 41.59 -----

15 29.71 0.99, 1.63 27.66 1.10, 1.72

16 27.48 1.61, 1.92 22.91 1.61, 1.97

17 55.24 ------46.47 ------

18 41.78 2.51 40.98 2.81

19 47.64 1.15, 1.63 45.84 1.16, 1.63

20 30.67 ------30.66 ------

21 39.43 1.22, 1.23 33.77 1.22, 1.33

22 31.45 1.57, 1.63 32.41 1.58, 1.77

23 18.43 0.87 28.08 0.98 s

24 18.32 0.78 15.52 0.75 s

25 23.46 0.91 15.52 0.91 s

26 17.18 0.81 17.06 0.77 s

27 23.63 1.21 25.91 1.13 s

28 172.93 ------182.66 ------

61

29 27.19 0.89 33.05 0.92 s

30 27.48 0.89 23.66 0.90 s

3.5.3. Physical properties of compound VK123

Physical state: white powder

Yield: 220 mg

Melting point: 233 oC

Molecular weight: 473.0778

IR vmax: OH stretch at 3444, C-H stretch at 2942 & 2895, C=O at 1695, ar C=C at 1468,

(COOH) at 1746, C-O at 1032cm-1.

30 29

19 20 21 12 18 17 22 11 OH 25 26 13 1 HO 9 14 16 28 2 O 10 8 15 3 5 4 HO 7 27 MA 6 24 23

Figure 3. 8: Structure of Maslinic acid. Table 3. 4: 1H and 13C-NMR data of Compound VK123 compared with literature data

26,27 27 Position δCMA δHMA δCLit δHLit

1 46.01 0.92, 1.99 46.2 0.91: 1.99

2 68.47 3.21(m) 68.3 3.70(m)

3 83.46 3.09 (d,12 Hz) 82.3 3.01 (d, 9.5 Hz)

4 39.10 ------39.1 -----

62

5 55.01 0.85 55.1 0.85

6 18.12 1.36, 1.55 18.27 1.40, 1.55

7 32.73 1.23, 1.34 32.59 1.32, 1.46

8 39.05 ---- 39.23 -----

9 47.45 1.62 47.6 1.62

10 38.03 ------37.05 -----

11 23.25 2.04:1.79 23.37 0.90, 1.95

12 121.92 5.35(t, 4 Hz) 122.61 5.30(t, 3.65 Hz)

13 143.71 ----- 143.26 -----

14 41.63 ----- 41.59 -----

15 27.4 1.09, 1.61 27.66 1.09, 1.71

16 23.0 1.62, 1.99 22.91 1.62, 1.99

17 46.43 ------46.47 ------

18 41.0 2.51 40.98 2.88

19 45.74 1.63, 1.65 45.84 1.63, 1.66

20 30.67 ------30.66 ------

21 33.64 1.22, 1.33 33.77 1.22, 1.35

22 31.45 1.57, 1.60 32.41 1.59, 1.78

23 28.3 0.80 28.08 1.03 s

24 16.67 0.83 15.52 0.83 s

25 16.48 0.99 15.52 0.99 s

26 16.18 0.90 17.06 0.77 s

27 23.2 1.15 25.91 1.14 s

28 178.5 ------183.66 ------

29 32.71 0.85 33.05 0.91 s

63

30 23.147 0.85 23.28 0.93 s

3.6. Discussion

3.6.1. Compound (VK121)

The Rf value of compound VK121 was determined using a TLC plate and hexane-ethyl acetate (7:3 v/v) as mobile phase. The Rf value (0.75) of VK121 was calculated by the formula described in Figure 3.5.

―FT-IR spectrum of compound VK121 (figure 3.9 APPENDIX ONE) indicated a broad peak

1 -1 2 around 3536-3469 cm- (OH stretch) and two aliphatic peaks at 3084-3012 cm (-CH sp

-1 3 -1 stretch), 2926-2856 cm (-CH sp stretch), 1513-1203 cm (-C=C, aromatic stretch), 1610 cm-1 (C=C alkene), 1267 cm-1 (C-O ether) and 1034 cm-1 (C-O alcohol).

For NMR analysis, compound VK121 was dissolved in spectroscopic grade deuterated

1 13 1 chloroform (CDCl3) to record H & C NMR Spectra. H NMR spectrum (figure 3.10,

APPENDIX ONE) showed various signals with OH proton at  5.01 (1H, s), aromatic protons ring at 6.40 (1H, s), 6.45 (1H, d), 6.50 (1H, d), two doublets at 3.22 (2H, d), 4.93 and 4.96 (1H, each d), a singlet with three protons at 3.73 (3H, s) which belongs to the methoxy group (-OCH), and a multiplet with one proton at 6.30 (1H, m).

13C NMR spectrum (figure 3.11, APPENDIX ONE) indicated 10 different signals which correspond to 10 carbon atoms. It indicated a signal at 142.7 ppm for C attached to hydroxyl

(OH), five aromatic carbons at 116.8, 122.8, 131.1, 114, 151.3, alkane carbon at 48, 56.2 (C-

O), and two carbons for alkene at 136.5, 117.2. The complete 1H and 13C NMR data is given in Table 3.2 which correlated well with the literature reported by Kumar L 2010 thus

Compound VK121 was identified as eugenol. The structure of eugenol is shown in Figure

64

3.6. The LC-MS results =264.0616 in figure 3.12 APPENDIX ONE are in good agreement with the theoretical molecular weight of eugenol.‖

3.6.2. Compound (VK122)

―Compound (VK122) was obtained as white powder with Rf value of 0.45, and a m.p. of 240 oC. The FT-IR spectrum (Figure 3.13, APPENDIX TWO) showed the presence of hydroxyl group (-OH) at 3443 cm-1, a carbonyl group at 1693 cm-1(-C=O), 2941 - 2870 cm-1 (-CH),

1468 cm-1 (-C=C) and 1031-998 cm-1 (-C-O).

The 1H NMR spectrum (figure 3.14, APPENDIX TWO) of compound VK122 showed signals of an olefinic proton at 5.35 cm-1 and seven methyls at 0.87, 0.78, 0.91, 0.81, 1.21,

0.89 and 0.89 which confirmed the characteristics of the oleanane skeleton28. This structural type was further supported by the 13C NMR spectrum (figure 3.15, APPENDIX TWO), which also contained resonance for olefinic carbons at (143.22 and 123.07 ppm) and one oxygenated carbons at 79.03 ppm and a carboxylic acid at 172.93 ppm. Compound VK122 was identified as oleanolic acid. The complete 1H and 13C NMR data is given in Table 3.3 and APPENDIX TWO, Full characterization of compound VK122 was done by comparison with literature reported data for oleanolic acid. The LC-MS results =456.3598 in figure 3.16

APPENDIX TWO are in good agreement with the theoretical molecular weight of oleanolic acid. The structure of VK122 is given in figure 3.7.‖

3.6.3. “Compound (VK123)

―Compound VK123 with Rf value of 0.14 was also isolated as a white powder. The spectral data of this compound were almost similar to that of compound VK122 however; 13C NMR

(figure 3.18, APPENDIX THREE) of VK123 indicated an extra oxygenated carbon signal at

68.47 ppm due to the presence of –OH group at C2 of its chemical structure. The FTIR

65 spectrum (figure 3.16, APPENDIX THREE) also indicted peaks of hydroxyl group (OH), carbonyl(C=O) and alkene at 3412, 1690 cm-1 and 1468 cm-1 respectively.

The 13C NMR spectrum exhibited signals for 30 carbon atoms with one carbonyl at 178.4 ppm, two olefinic carbons at (121.92 and 143.71). The 1H NMR spectrum (figure 3.16,

APPENDIX THREE) showed an olefinic proton signal at 5.50 cm-1. The complete 1H and 13C

NMR data is given in Table 3.3 and APPENDIX THREE. Compound VK123 was identified as maslinic acid by comparison of the obtained 1H and 13C NMR spectra with those of maslinic acid reported in literature and LC-MS confirmed the molecular weight=473.0778.

The structure of VK123 is given in figure 3.8.‖

66

3.7. References

1. Cortés-Rojas DF, Souza CRF de, Oliveira WP, Laboratory. Clove ( Syzygium

aromaticum ): a precious spice. Asian Pac J Trop Biomed. 2014;4(2):90-96.

doi:10.1016/S2221-1691(14)60215-X

2. Bhowmik D, Kumar KPS, Yadav A, Srivastava S, Paswan S, Sankar A. Recent Trends

in Indian Traditional Herbs Syzygium aromaticum and its Health Benefits. J

Pharmacogn Phytochem. 2012;1(1):13-22.

3. Ajiboye TO, Mohammed AO, Bello SA, Yusuf II, Ibitoye OB, H.F. Muritala HF,

Onajobi IB . Antibacterial activity of Syzygium aromaticum seed : Studies on oxidative

stress biomarkers and membrane permeability. Microb Pathog. 2016;95:208-215.

doi:10.1016/j.micpath.2016.03.011

4. El-Maati MFA, Mahgoub SA, Labib SM, Al-Gaby AMA, Ramadan MF. Phenolic

extracts of clove (Syzygium aromaticum) with novel antioxidant and antibacterial

activities. Eur J Integr Med. 2016;8(4):494-504. doi:10.1016/j.eujim.2016.02.006

5. Aisha AFA, Abu-salah KM, Salman A. Syzygium aromaticum extracts as good source

of betulinic acid and potential anti-breast cancer. Brazilian J Pharmacogn.

2012;22(2):335-343.

6. Orwa C, Mutua A, Kindt R, Jamnadass R SA. Syzygium aromaticum ( L .) Merr . &

Perr . Agrofor Database. 2009;4.0:1-5.

7. Wankhede TB. Evaluation of antioxidant and antimicrobial activity of the Indian. Int

Res J Sci Eng. 2015;3(4):166-172.

67

8. Shrivastava K, Sahu S, Mishra SK, De K. In vitro Antimicrobial Activity and

Phytochemical Screening of Syzygium aromaticum. Asian J Pharm Sci. 2014;4(1):12-

15.

9. Pandey A, Singh P. Antibacterial activity of Syzygium aromaticum (clove) with metal

ion effect against food borne pathogens. Asian J Plant Sci Res. 2011;1(2):69-80.

10. Mbaveng AT, Kuete V. Syzygium Aromaticum. Elsevier Inc.; 2017. doi:10.1016/B978-

0-12-809286-6/00029-7

11. Parle M, Deepa K. Clove : A champion spice. Int J Res Ayurveda Pharm.

2016;2(1):47-54.

12. Devi KP, Nisha SA, Sakthivel R, Pandian SK. Eugenol ( an essential oil of clove ) acts

as an antibacterial agent against Salmonella typhi by disrupting the cellular membrane.

J Ethnopharmacol. 2010;130(1):107-115. doi:10.1016/j.jep.2010.04.025

13. Venugopal K, Rather HA, Rajagopal K, Shanthi MP,Sheriffc K, Illiyas M, Rather RA ,

Manikandan E, Uvarajan S, Bhaskar M, Maaza M. Synthesis of silver nanoparticles (

Ag NPs ) for anticancer activities ( MCF 7 breast and A549 lung cell lines ) of the

crude extract of Syzygium aromaticum. J Photochem Photobiol B Biol. 2017;167:282-

289. doi:10.1016/j.jphotobiol.2016.12.013

14. Sobeh M, Esmat A, Petruk G, Abdelfattah MAO, Dmirieh M, Montic DM, Abdel-

Naim AB, Wink M. Phenolic compounds from Syzygium jambos ( Myrtaceae ) exhibit

distinct antioxidant and hepatoprotective activities in vivo. J Funct Foods.

2018;41:223-231. doi:10.1016/j.jff.2017.12.055

15. Wang Y, Jia J, Tian Y, Shua X, Rena X, Guana Y, Yan Z. LWT - Food Science and

Technology Antifungal effects of clove oil microcapsule on meat products. LWT -

68

Food Sci Technol. 2018;89:604-609. doi:10.1016/j.lwt.2017.11.042

16. Hasheminejad N, Khodaiyan F, Safari M. Improving the antifungal activity of clove

essential oil encapsulated by chitosan nanoparticles. Food Chem. 2019;275:113-122.

doi:10.1016/j.foodchem.2018.09.085

17. Shukri R, Mohamed S, Mohamed N. Cloves protect the heart , liver and lens of

diabetic rats. Food Chem. 2010;122(4):1116-1121.

doi:10.1016/j.foodchem.2010.03.094

18. Ramadan BMF, Asker MMS, Tadros M. Lipid profile , antiradical power and

antimicrobial properties of Syzygium aromaticum oil. Grasas Y aceites.

2013;64(5):509-520.

19. Bao L-M, Eerdunbayaer, Nozaki A, Takahashi E, Okamoto K, Ito H, Hatanoa T.

Hydrolysable tannins isolated from syzygium aromaticum: structure of a new c-

glucosidic ellagitannin and spectral features of tannins with a tergalloyl group.

Heterocyclic. 2012;85(2):365-381. doi:10.3987/COM-11-12392

20. Kamatou GP, Vermaak I, Viljoen AM. Eugenol—From the Remote Maluku Islands to

the International Market Place: A Review of a Remarkable and Versatile Molecule.

Molecules. 2012;17(6):6953-6981. doi:10.3390/molecules17066953

21. Pino JA, Marbot R, Aguero J. Essential Oil from Buds and Leaves of Clove (Syzygium

aromaticum ( L .) Merr . et Perry) Grown in Cuba. J Essent Oil Res. 2001;13:278-279.

22. Rali S, Oyedeji OO, Aremu OO, Oyedeji AO, Nkeh-Chungag BN. Semisynthesis of

derivatives of oleanolic acid from Syzygium aromaticum and their antinociceptive and

anti-inflammatory properties. Mediators Inflamm. 2016;2016:1-9.

doi:10.1155/2016/8401843

69

23. Taganna JC, Quanico JP, Perono RMG, Amor EC, Rivera WL. Tannin-rich fraction

from Terminalia catappa inhibits quorum sensing (QS) in Chromobacterium

violaceum and the QS-controlled biofilm maturation and LasA staphylolytic activity in

Pseudomonas aeruginosa. J Ethnopharmacol. 2011;134(3):865-871.

doi:10.1016/j.jep.2011.01.028

24. Kumar L Tyagi P. Antifungal , antibacterial and insect repellent chemical molecules of

ocimum sanctum l, PhD thesis, Maharaj Singh College Saharanpur (India); 2010.

25. Hichri F, Ben H, Cheriaa J, Jegham S, Mighri Z. Antibacterial activities of a few

prepared derivatives of oleanolic acid and of other natural triterpenic compounds.

Comptes Rendus Chim. 2003;6:473-483. doi:10.1016/S1631-0748(03)00066-3

26. Khathi A, Serumula MR, Myburg RB, Van HFR, Musabayane CT. Effects of

Syzygium aromaticum -Derived Triterpenes on Postprandial Blood Glucose in

Streptozotocin-Induced Diabetic Rats Following Carbohydrate Challenge. PLoS One.

2013;8(11):1-8. doi:10.1371/journal.pone.0081632

27. Dais P, Plessel R, Williamson K, Hatzakis E. Complete 1H and 13C-NMR assignment and 31P-NMR determination of pentacyclic triterpenic acids. Anal Methods. 2017;9:949-957. doi:10.1039/c6ay02565j

28. Hossain MA, Zhari I. Isolation and characterization of triterpenes from the leaves of Orthosiphon stamineus. Arab J Chem. 2013;6(3):295-298.

70

CHAPTER FOUR

SYNTHESIS OF OLEANOLIC ACID ANALOGS

4.1. Materials

―All chemicals and―reagents used were purchased from Sigma-Aldrich and used without further purification. The solvents used for synthesis were of high grade solvents and they were dried over molecular sieves with pore size of 4 Å and particle size of 4-8 Mesh.‖

4.2. Characterization

―The fingerprint identification of specific functional groups was achieved by assigning of group frequencies (cm-1) of particular peaks within the IR spectra. The IR spectra were obtained from

Perkin Elmer model 100 Hz and the percentage (%) transmittance was recorded against the wave length (cm-1) for the reported signals in the range of 4000- 400 cm-1.

The 1H and 13C NMR spectra were recorded on Bruker Nuclear Magnetic Resonance

Spectrometer 400 MHz at room temperature using deuterated CDCl3. The chemical shifts (δ) are reported in ppm relative to internal solvent peaks and the coupling constants (J) were measured in Hertz (Hz). The 1H signals splitting pattern abbreviation were reported in the following order: singlet (s), (doublet), triplet (t), quartet (q), doublet of doublet (dd), and multiplet (m).

The high resolution mass spectra were recorded on Bruker Compact Liquid Chromatography

Mass Spectrometry (LC-MS).

71

Finally, the melting points (m.p) were determined on a variable heat Gallenkamp melting point apparatus (temperature range 50–350 °C) equipped with the laboratory thermometer.‖

4.3. Introduction

―The synthesis of hybrid compounds containing oleanolic acid and other pharmaceutical scaffolds was performed by first synthesizing 4-aminoquinoline derivatives by amination reaction of either amines or amino alcohols with 4.7-dichloroquinoline resulting in compounds with targeted functional groups. The synthesis of 4-aminoquinoline derivatives was carried out via a nucleophilic substitution on the suitably substituted 4.7-dichloroquinoline scaffold. This was achieved by using amines and amino alcohols as nucleophils.‖

R NH2

Cl H R Cl N R HN -H -Cl

Cl N Cl N Elimination of a leaving group Cl N H H neuclephilic addition B on Cloroquinolium ring

Scheme 4. 1: N-alkylation of 4.7-dichloroquinoline mechanism. The hybrid compounds containing oleanolic acid were then prepared from the reaction of oleanolic acid with isolated 4-aminoquinoline derivatives and other selected organic compounds such as curcumin or ergocalciferol via amidation or esterification reactions. The reactions were performed at 75-95oC and monitored by TLC plate to ensure the completion of reaction. The synthesized hybrid compounds were further purified by column chromatography and characterized by using FT-IR, NMR, m.p, and LC-MS.‖

72

4.3. Synthesis of 4-aminoquinoline derivatives.

―Chloroquine is a 4-aminoquinoline ―derivative with a wide range of pharmacological activities such as, anti-malarial1, anti-bacterial2, clonorchis sinesis, anti-fungal3, and rheumatoid arthritis.

Chloroquine derivatives with various alkyl amino side chain were synthesized by one-step substitution reaction of an appropriate amine and 4,7-dichloroquinoline. Some studies have proven that lengthening and shortening of the linker of the alkyl amine side chain in chloroquine leads to compounds that remain effective against drug-resistant strains4.‖

4.4. Synthesis of N-(3-aminopropyl)-7-chloroquinolin-4-amine (1.3 PDA-Q)

HN Cl H N NH Cl Cl 2 2 NH2 900C 24 hr N N N-(3-aminopropyl)-7-chloroquinolin-4-amine

Scheme 4. 2: synthesis of N-(3-aminopropyl)-7-chloroquinolin-4-amine. ―A solution of 4,7-dichloroquinoline (1 g, 5.05 mmol) in 1,3-diaminopropane(1.3 PDA) (1.9 mL, 22.7 mmol) was refluxed for 24 hours at 90 oC with stirring, and the reaction was monitored by TLC using methanol: Triethanolamine (TEA): hexane (6:3:1) as eluent solvent system to ensure the completion of reaction, then allowed to cool at room temperature. The mixture was diluted with 60 mL dichloromethane (DCM) (20 mL x 3) and the resulting mixture was successfully washed with sodium hydroxide (NaOH) (1 M, 10 mL x 3) followed by 10 mL brine, to give an aqueous layer, an organic layer, and a white course particulate precipitate. The organic layer was filtered through a cotton plug and concentrated in a rotary evaporator. N-(3-

o aminopropyl)-7-chloroquinolin-4-amine (Yield 62 %, m.p 129-131 C, Rf value 0.34) was

73 obtained as pale yellow crystals. The physical characteristics were almost the same as previously described by Sunduru et al., 20095.

The FTIR spectrum showed the successful synthesis of N-(3-aminopropyl)-7-chloroquinolin-4- amine by indicating the peaks of N-H stretch for secondary and primary amine at 3357 cm-1,

3279 cm-1 respectively, CH stretch at 2947 cm-1, C-Cl stretch at 601 cm-1 and C=C stretch for aromatic at 1582 cm-1 .‖

95

85

75

65 Transmitance % 55

45

35 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 4. 1: ―IR spectrum of N-(3-aminopropyl)-7-chloroquinolin-4-amine.”

74

4.5. Synthesis of N-(2-aminopropyl)-7-chloroquinolin-4-amine (1.2 PDA-Q)

Cl HN H2N NH NH2 2

0 Cl N 90 C 24 hr Cl N

N-(2-aminopropyl)-7-chloroquinolin-4-amine

Scheme 4. 3: Synthesis of N-(2-aminopropyl)-7-chloroquinolin-4-amine. ―A mixture of 4,7-dichloroquinoline (1 g, 0.05 mmol) and 1,2-diaminopropane (1.9 mL, 22.7 mmol) was heated to reflux at 90 oC for 24 hours with stirring, and the reaction was monitored by TLC using MeOH: TEA: EtOAc (6:3:1) as eluent solvent system to ensure the completion of reaction, then allowed to cool to room temperature. The mixture was diluted with 60 mL DCM

(20 mL x 3) and the resulting mixture was successfully washed with sodium hydroxide (NaOH)

(1 M, 10 mL) and brine (10 mL), to give an aqueous layer, an organic layer, and a white course particulate precipitate. The organic layer was filtered through a cotton plug and concentrated in a rotary evaporator. N-(2-aminopropyl)-7-chloroquinolin-4-amine (Yield 66 %, m.p 128–130 oC,

Rf value 0.64) was obtained as pale yellow crystals.

The FTIR spectra for N-(2-aminopropyl)-7-chloroquinolin-4-amine molecule, peaks that were observed are N−H stretch visible at 3366 cm-1, C−H stretch alkane at 2901 cm-1, C=C stretch aromatic at 1587 cm-1 and C−Cl stretch at 756.5 cm-1 respectively which confirmed the successful linkage of ethyl diamine to the quinoline moiety.‖

75

EDAQ 100

90

80

70

60

Transmittance (%) Transmittance 50

40

30

4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 4. 2: IR spectrum of N-(2-aminopropyl)-7-chloroquinolin-4-amine (1.2 PDA-Q)

4.6. Synthesis of N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine (DET.Q)

H Cl N H HN NH2 N H2N NH2 850C 24 h Cl N Cl N N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine

Scheme 4. 4: Synthesis of N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine

76

―N-(2-aminopropyl)-7-chloroquinolin-4-amine was prepared using a modified procedure described by Musonda at al 20066. A solution of 4,7-dichloroquinoline (1.00 g, 5.05 mmol) in diethelenetriamine (DET) (1.05 g, 10.1 mmol) was heated to reflux at 85°C for 24 hours with stirring, the reaction was monitored by TLC using MeOH: TEA: Hex (6:3:2) as eluent solvent system to ensure the completion of reaction, then allowed to cool to room temperature. The mixture was basified with 20 ml 1 M NaOH, extracted with 60 ml hot EtOAc (20 ml x 3) and the resulting mixture was successfully washed with Na2SO4 (10 ml). The separated organic layer was filtered and concentrated in a rotary evaporator. N-(2-aminopropyl)-7-chloroquinolin-4-

0 amine (Yield 85 %, m.p 112–114 C, Rf value 0.56) was obtained as yellow crystals.

The FTIR spectra showed a successful synthesis of N-(2-(2-Aminoethylamino) ethyl)-7- chloroquinoline-4-amine (DET-Q) by showing the visible peaks of N-H stretch at 3302 cm-1, C-

H stretch for alkene at 3005 cm-1, C-H stretch at 2947, cm-1, C=C for the aromatic at 1579 cm-1 and C-Cl at 647 cm-1 . The obtained IR results were in good agreement with results reported in literature6,7.‖

100

90

80

70

60

50

40 Transmitance %

30

20 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1 Figure 4. 3: IR spectra of N-(2-(2-Aminoethylamino) ethyl)-7-chloroquinoline-4-amine.

77

4.7. Symthesis of 1-(7-Choloroquinolin-4-yl) hydrazine (HYD-Q)

NH2 Cl HN

H2N NH2 950C 24 h Cl N Cl N 1-(7-Choloroquinolin-4-yl) hydrazine

Scheme 4. 5: Synthesis of 1-(7-Choloroquinolin-4-yl) hydrazine (Hyd-Q)

A solution of 4.7- dichloroquinoline (1g, 5. 05 mmole) and Hydrazine (0.75 ml, 30.30 mmole) in

3 ml ethanol as solvent was refluxed at 95 oC for 24 hr. the reaction was monitored by TLC using toluene: EtOAc: meOH (6:3:1) as eluent solvent system to ensure the completion of reaction, the obtained solution was filtered and allowed to dry in fume-cardboard then later it was recrystallized with 10 ml ethanol. 1-(7-Choloroquinolin-4-yl) hydrazine (Yield 72 %, m.p 129–

0 132 C, Rf value 0.64) was obtained as pale yellow crystals. The FTIR spectrum shown in figure

4.4 confirmed a successful synthesis of 1-(7-Choloroquinolin-4-yl) hydrazine by indicating N-H stretch for secondary amine at 3278 cm-1, C-H stretch for alkene at 3120 cm-1, C-H stretch for alkane at 2945 cm-1, C=C 1550 cm-1 for aromatic and also C-Cl for alkyl halide at 643 cm-1. The obtained results of 1-(7-Choloroquinolin-4-yl) hydrazine were in good agreement with results reported by Pretorius et al 20138.

78

90 85 80

75 70 65 60

55 transmitance% 50 45 40 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 4. 4: FT-IR spectrum of 1-(7-Choloroquinolin-4-yl) hydrazine

4.8. Synthesis of 2-(7-chloroquinolin-4-ylamino) ethanol (EA-Q)

OH Cl HN OH H2N 850C 24 h Cl N Cl N 2-(7-chloroquinolin-4-ylamino)ethanol

Scheme 4. 6: Synthesis of 2-(7-chloroquinolin-4-ylamino) ethanol (EA-Q)

―4,7-dichloroquinoline (1000 mg, 5.05mmol) was dissolved in EA (3.05ml). The reaction was refluxed over a period of 24 hours at 85oC. the reaction was monitored by TLC using toluene:

EtOAc: MeOH (6:3:1) as eluent solvent system to ensure the completion of reaction, After 24 hours, the reaction was allowed to cool at room temperature and 30ml of distilled water was added into the solution and the resultant was filtered, the solids were then allowed to dry at room temperature. Then later it was recrystallized with 20 ml of methanol. 2-(7-chloroquinolin-4- ylamino) ethanol (Yield 65%, m.p 112–114 oC, Rf value 0.72) was obtained as pale yellow crystals.

79

The FTIR results confirmed a successful incorporation of 4.7 dichloroquine and ethanolamine

(EA), it indicated N-H stretch for secondary amine at 3300 cm-1, O-H stretch for alcohol at 3190 cm-1, C-H stretch for alkene at 3111 cm-1, also for C-H stretch for alkane at 2802 cm-1, C=C for aromatic at 1550 cm-1, C-N stretch for amine at 1083 cm-1 and also C-Cl at 750 cm-1. The above peaks confirmed the successful isolation of compound 2-(7-chloroquinolin-4-ylamino) ethanol. 9

90

80

70

60 Transmitance Transmitance %

50

40 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1 Figure 4. 5: FT-IR spectrum of 2-(7-chloroquinolin-4-ylamino) ethanol

80

4.9. Synthesis of 2-(2-(-chloroquinolin-4-ylamino)ethoxy)ethanol (AEE-Q)

O Cl HN OH O H2N OH 850C 24 h Cl N Cl N 2-(2-(7-chloroquinolin-4-ylamino)ethoxy)ethanol

Scheme 4. 7: Synthesis of 2-(2-(-chloroquinolin-4-ylamino)ethoxy)ethanol (AEE-Q)

―4,7-dichloroquinoline (1000 mg, 5.05mmol) was dissolved in AEE (3.05ml. The reaction was refluxed over a period of 24 hours at 85oC. After 24 hours, the reaction was allowed to cool at room temperature and 30ml of distilled water was poured into it and the resultant was filtered, the solids were dried at room temperature, 1ml of methanol and 5ml of ethyl acetate was used to boil those solids and ice was used to cool them after solids were collected. 2-(2-(-chloroquinolin-

4-ylamino)ethoxy)ethanol (Yield 75%, m.p 122–124 oC, Rf value 0.62) was obtained as brownish orange crystals. The FTIR revealed the presence of the expected functional groups in

AEE-Q. it revealed N-H stretch for secondary amine at 3310 cm-1, C-H stretch for alkane at 2803 cm-1, C=C for aromatic at 1580 cm-1, C-N for amine at 1108 cm-1, C-O for ester at 1050 cm-1 and

-1 9 also C-Cl at 530 cm . (6:2:2 methanol/TEA/Hexane, Rf = 0.54) .‖

81

95

85

75

65

55 Transittance %

45

35 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 4. 6: FT-IR spectrum of 2-(2-(-chloroquinolin-4-ylamino)ethoxy)ethanol (AEE-Q).

4.10. Synthesis of 4-(7-chloroquinolin-4-ylamino)-2-hydroxybenzoic acid

O O OH Cl OH HN OH

H2N OH

DMSO 850C 24 hrs Cl N Cl N 4-(7-chloroquinolin-4-ylamino)-2-hydroxybenzoic acid

Scheme 4. 8: Synthesis of 4-(7-chloroquinolin-4-ylamino)-2-hydroxybenzoic acid ―4-aminosalicylic acid (70 mg, 0.45 mmol) was dissolved in 5 mL DMSO followed by addition of 4.7-dichloroquinoline (100 mg, 0.45 mmol). The reaction was allowed to stir for approximately 10 minutes until all the solute were completely dissolved followed by the addition of DMAP (55 mg, 0.45 mmol). The reaction was allowed to stir for 10 minutes followed by the addition of DCC (103 mg, 0.50 mmol) in portion within a period of 5 minutes. The reaction was

82 allowed to stir overnight at 85oC temperature. It was monitored by TLC using (6:4 toluene/ethyl acetate, and Rf = 0.24). The obtained product was extracted three times using 20 mL dichloromethane and 20 mL cold distilled water. The organic layer was dried over anhydrous sodium sulphate, filtered and then concentrated on the roti- evaporator. A viscous liquid was obtained which was further purified by column chromatography (6:4:1 Toluene/Ethyl acetate/Methanol). (0.108 g), Yield: (68%). FT-IR (cm-1): 3382 (N−H), 2981 (C−H), 15621

(C=C aromatic), 1695 (C=O), 1288 (N−H bending) and 1176 (C−O)10.‖ PDAQ 100

90

80

70

60

50

Transmittance (%) Transmittance 40

30

20

10 4000 3500 3000 2500 2000 1500 1000 500

Wavenumber cm-1

Figure 4. 7: FT-IR spectrum of 4-(7-chloroquinolin-4-ylamino)-2-hydroxybenzoic acid

83

―The first step of the proposed strategy was to synthesise chloroquine derivatives with various alkyl amino side chain. The synthesis was carried out via a nucleophilic substitution reaction on the suitably substituted 4,7-dichloroquinoline scaffold. This was successfully achieved by using amine or amino alcohol as nucleophiles.

―According to the reported experimental methods in literature2,6,11, a mixture of chloroquine and amine is commonly homogenised at 80 oC, followed by reflux at high temperature(i.e 120-140 oC). Similarly, the experiments were refluxed at 120 °C and monitored by TLC plate. However, the reactions at 120 °C resulted in decomposition of the precursors.

From this observation, it was concluded that the high temperatures became unfavourable for the reactions. Decomposition has been confirmed as the major problem for similar reactions and milder reaction conditions have been considered useful for these reactions to be successful, i.e. reactions at 75-95°C rather than high temperatures12. All the synthesised 4-aminoquinoline derivatives were characterized with FT-IR which indicated the successful isolation of compounds as reported by some researchers.‖

84

4.11. DERIVATIVES OF OLEANOLIC ACID.

4.11.1. Synthesis of hybrid compounds with amide linkers

4.11.2 Synthesis of compound VK1.

Cl N Cl N

H OH N NH H2N NH O O HO HSU, DCC, ref 120 0C HO Scheme 4. 9: Synthesis of compound VK1

39 N 35 Cl ―Oleanolic acid (200 mg, 0.4mmol) and 1.3 23 30 40 36 34 43 21 22 20 41 37 33 42 38 PDA.Q (103 mg, 0.4mmol) was dissolved in 5 13 18 19 H 14 12 17 N 46 NH 25 24 27 48 47 45 44 6 10 11 16 mL of DMSO. N-Hydroxysuccinimide (HSU) 1 5 9 15 O31 2 4 8 29 HO 3 7 VK1 (50.40mg, 0.4mmol) and N,N‘- 32 28 26 Dicyclohexylcarbodiimide (DCC) (99.39 mg, 0.5 mmol) portions were added and stirred at room temperature for 10 minutes then heated to reflux at 120 0C for two days, the reaction was monitored by TLC to ensure the completion of reaction, after which the resultant solution was diluted with DCM (20 mL) and the mixture was washed with 30 mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulphate, filtered and concentrated on the rotary evaporator to obtain a light brown powder. The crude compound was purified by column chromatography using silica gel (Merck, silica gel 60 F254: 0.063-0.200 mm). The column was eluted with EtOAc: MeOH (9:1) to afford a

0 white powder (Yield 54%, m.p 131–133 C, Rf value 0.42). IR vmax 3296 (NH), 3076 (OH),

1 2932-2873 (aliphatic CH), 1626(C=O), 1545(C=C), 1337(C-N). H NMR (400 MHz, CDCl3): δ

(ppm) 8.28-8.26 (d, J=8.0 Hz, 1H, H-40), 7.32 (s, 1H, C-48), 7.18-7.16 (d, j=8.0 Hz, 1H, H-38),

85

7.08-7.06 (d, j=8.0 Hz, 1H, H-33), 5.66-5.64 (d, j=8.0 Hz, 1H, H-41), 5.28 (t, J=4.0 Hz, 1H, H-

13) 4.06-4.00 (q, j=8.0 Hz, 3H, H-2 & 47), 3.39(s, 1H, H-48), 3.17-3.13 (dd, j=4.0 Hz, 2H, H-

13 45), 2.48-2.43 (dd, j=4.0 Hz, 1H, H-18), 2.19 (s, 1H, H-32). C NMR (400 MHz, CDCl3): δ

(ppm) 177.13 (C27), 155.23 (C42), 152.43 (C40), 148.21 (C36), 144.73 (C12), 135.43 (C34),

129.11 (C35), 125.77 (C33), 122.53 (C38), 121.34 (C13), 119.41 (C37), 114.24 (C41), 78.97

+ + (C2). MS (ESI ) 673.44 calculated for C42H60ClN3O2 [M+H] , found 673.33.‖

4.11.3. Synthesis of compound VK2

N Cl N Cl

OH H NH N NH O H2N O HO HSU, DCC, ref 120 0C HO

Scheme 4. 10: Synthesis of compound VK2

38 N 34 Cl ―Oleanolic acid (400 mg, 0.8 mmol) and 23 29 39 35 33 42 21 22 20 40 36 32 41 37 HYD.Q (200 mg, 0.8 mmol) was dissolved in 18 13 19 H 14 12 17 N NH 24 26 44 43 5 mL of DMSO. HSU (100.8 mg, 0.8 mmol) 6 10 11 16 1 5 9 15 O

2 4 8 28 VK2 and DCC (99.39 mg, 0.5 mmol) portions were HO 3 7 31 27 25 added and stirred at room temperature for 10 minutes then heated to reflux at 120 0C for 24 hr, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with DCM (20 mL) and the mixture was washed with 30 mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulphate, filtered and

86 concentrated on the rotary evaporator to obtain off-white crystals. The crude was purified by column chromatography using silica gel (Merck, silica gel 60 F254: 0.063-0.200 mm). The column was eluted with EtOAc: Hex (7:3) to afford a white powder (Yield 66 %, m.p 114–115 o C, Rf value 0.33). IR vmax 3548 (NH), 3383 (OH), 2929-2857 (CH aliphatic), 1709(C=O),

1 1631(C=C alkene), 1455(C=C aromatic), 1217(C-N), 628 (C-Cl). H NMR (400 MHz, CDCl3):

δ (ppm) 8.06-8.04 (d, J=8.0 Hz, 1H, H-39),7.89(s,1H, C34), 7.62-760 (d, J=8.0 Hz, 1H, C-37),

7.44-7.42 (d, J= 8.0 Hz, 1H, H-32), 5.65-5.63 (d, J=8.0 Hz, 1H, H-40), 5.28 (t, J=4.0 Hz 1H, H-

13), 3.40 (s, 1H, H-44), 3.17-3.13 (dd, J=4.0 Hz, 1H, H-2), 2.48-2.43 (dd, J=4.0 Hz, 1H, H-18),

13 2.10 (s, 1H, H-31). C NMR (400 MHz, CDCl3): δ (ppm) 181.52 (C26), 152.12 (C39), 149.97

(C41), 148.24 (C35), 144.74 (C12), 135.43 (C33), 129.65 (C34), 127.61 (C32), 122.52 (C37),

121.99 (C13), 118.42 (C36), 113.65 (C40), 78.97 (C2). MS (ESI+) 631.39 calculated for

+ C39H54ClN3O2 [M+H] , found 631.35.‖

87

4.11.4. Synthesis of compound VK3

N

H2N OH N H N H N Cl N O H O HO HSU, DCC, ref 120 0C Cl HO Scheme 4. 11: Synthesis of compound VK3

34 27 ―Oleanolic acid (200 mg, 0.4 mmol) 25 26 24 44 45 N 43 17 22 23 and 1.2 PDA.Q (103 mg, 0.4 mmol) H 18 16 21 N 1 46 40 29 31 4 2 N 41 39 10 14 15 20 H 48 5 9 13 19 42 38 was dissolved in 5 mL of DMSO. HSU O 3 37 Cl 6 8 12 47 HO 7 11 36 VK3 (50.40 mg, 0.4 mmol) and DCC (99.39 32 30 mg, 0.5 mmol) portions were added and stirred at room temperature for 10 minutes then heated to reflux at 120 0C for 24 hours, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with DCM (20 mL) and the mixture was washed with 30 mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over sodium sulphate, filtered and concentrated on the rotary evaporator to obtain a white powder. The crude was purified by column chromatography using silica gel

(Merck, silica gel 60 F254: 0.063-0.200 mm). The column was eluted with EtOAc: Hex (7:3) to

0 afford a white powder (Yield 51%, m.p 122–124 C, rf value 0.52). IR vmax 3546 (NH), 3359

(OH), 2928-2857 (CH aliphatic), 1712 (C=O), 1637(C=C alkene), 1456 (C=C aromatic),

1 1223(C-N), 626 (C-Cl). H NMR (400 MHz, CDCl3): δ (ppm) 7.92 (s, 1H, H-4), 7.62 (s, 1H, H-

39), 7.56-7.54 (d, J= 8.0 Hz, 1H, H-42), 7.41-7.39 (d, J=8.0 Hz, 1H, H-37), 5.64-5.62 (d, J=8.0

Hz, 1H, H-45), 5.28 (t, J=4.0 Hz 1H, H-17), 4.45 (s, 1H, H-48), 4.32-4.30 (d, J=8.0 Hz, 1H, H-

88

2), 3. 17- 3.13 (dd, J=4.0 Hz, 1H, H-6), 2.48- 2.44(dd, J=4.0 Hz, 1H, H-22), 2.19 (s, 1H, H-36).

13 C NMR (400 MHz, CDCl3): δ (ppm) 177.03 (C31), 155.75 (C38), 150.23 (C42), 148.23 (C40),

144.00 (C16), 133.75 (C46), 128.43 (C47), 126.41 (C45), 122.50 (C44), 121.13 (C17), 119.23

+ + (C39), 114.34 (C43), 78.96 (C6). MS (ESI ) 673.44 calculated for C42H60ClN3O2 [M+H] , found

671.09.‖

4.11.5. Synthesis of compound VK4

N Cl N Cl

OH H H N NH N NH 2 N N O H H O HO 0 HSU, DCC, ref 120 C HO Scheme 4. 12: Synthesis of compound VK4

7 N 3 Cl ―Oleanolic acid (200 mg, 0.4 mmol) 48 41 8 4 2 11 39 40 38 9 5 1 10 6 and DET.Q (103 mg, 0.4 mmol) was 31 36 37 H 32 30 35 N 16 15 14 NH 43 42 45 18 17 N 13 12 dissolved in 5 mL of DMSO. HSU 24 28 29 34 H 19 23 27 33 O 49 20 22 26 47 (50.40 mg, 0.4 mmol) and DCC HO 21 25 VK4 50 46 44 (99.39 mg, 0.5 mmol) portions were added and stirred at room temperature for 10 minutes then heated to reflux at 120 0C for two days, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with DCM (20 mL) and the mixture was washed with 30 mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulphate, filtered and concentrated on the rotary evaporator to obtain a white powder. The crude compound was purified by column chromatography using silica gel

(Merck, silica gel 60 F254: 0.063-0.200 mm). The column was eluted with EtOAc: MeOH (9:1) to

89

0 afford a white powder (Yield 62%, m.p 128–130 C, rf value 0.52). IR vmax 3356 (NH), 3265

(OH), 2943-2857 (CH aliphatic), 1712 (C=O), 1637(C=C alkene), 1456 (C=C aromatic),

1 1223(C-N), 626 (C-Cl). H NMR (400 MHz, CDCl3): δ (ppm) 7.74-7.72 (d, J=8.0 Hz, 1H, H-8),

7.14(s, 1H, H-3), 7.04-7.02(d, J=8.0 Hz, 1H, H-6), 6.74-6.72(d, j=8.0, 1H, H-1), 6.42-6.40(d,

J=8.0, 1H, H-9), 5.28 (t, J=4.0, 1H, H-31), 4.45 (s, 1H, H-12), 4.04 (m, 4H, H-13& H-17), 3.66

(q, J=8.0, 4H, H-14& 16), 3.40 (s, 1H, H-18), 3.14 (dd, J=8.0, 1H, H-36), 2.45 (dd, 4.0 Hz, 1H,

13 H-20), 1.98 (s, 1H, H-50). C NMR (400 MHz, CDCl3): δ (ppm) 177.07 (C45), 154.65 (C10),

151.30 (C8), 148.44 (C4), 144.75 (C30), 134.50 (C2), 129.13 (C3), 127.41 (C1), 122.50 (C6),

121.99 (C31), 119.09 (C5), 113.21 (C9), 78.96 (C20), 48.08 (C13), 47.57 (C17), 46.81 (C14),

+ + 46.23 (C16). MS (ESI ) 702.46 calculated for C43H63ClN3O2 [M+H] , found 701.38.‖

90

4.12. Synthesis of hybrid compounds with ester linkers.

4.12.1. Synthesis of compound VK5.

HO NH OH Cl

O N O NH HO O Cl DMSO, DCC,DMAP, reflux 120 0C 24 h HO N Scheme 4. 13: Synthesis of compound VK5

30 23 ―Oleanolic acid (400 mg, 0.8 mmol) and EA.Q 21 22 20

13 18 19 (195.02 mg, 0.8 mmol) was dissolved in 5 mL 14 12 17 O 34 25 24 27 33 34 36NH 6 10 11 16 of DMSO. 4-Dimethylaminopyridine (DMAP) 1 5 9 15 O31 37 46 Cl 38 42 45 47 2 4 8 29 HO 3 7 39 41 44 32 N 43 (97.74 mg, 0.8 mmol) and DCC (181.56 mg, 28 26 40 VK5 0.88 mmol) portions were added and stirred at room temperature for 10 minutes then heated to reflux at 120 0C for 28 hours, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with DCM (20 mL) and the mixture was washed with 30mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulfate, filtered and concentrated on the rotary evaporator to obtain a light brown powder. The crude was purified by column chromatography using silica gel (Merck, silica gel 60

F254: 0.063-0.200 mm). The column was eluted with EtOAc: MeOH (9:1) to afford white crystals

0 (Yield 54%, mp 131–133 C, Rf value 0.43).

IR vmax 3450 (NH), 3307 (OH), 2943-2897 (CH aliphatic), 1694 (C=O), 1468(C=C alkene), 1398

1 (C=C aromatic), 1274(C-N), 660 (C-Cl). H NMR (400 MHz, CDCl3): δ (ppm) 7.94-7.92 (d,

91

J=8.0 Hz, 1H, H-39), 7.78(s, 1H, H-46), 7.58-7.56 (d, J=8.0 Hz, 1H, H-44), 5.64-5.62(d, J=8.0

Hz, 1H, H-38), 5.38(t, J=4.0 Hz,1H, H-13) 4.45(s, 1H, H-36), 4.34-4.29(dd, J=8.0 Hz, 1H, H-

34), 4.05-4.00 (dd, J=8.0 Hz, 2H, H-35), 3.17-3.13(dd, J=4.0 Hz, 1H, H-2), 2.48-2.44(dd, J=4.0

Hz, 1H, H-18).

13 C NMR (400 MHz, CDCl3): δ (ppm) 177.05 (C27), 152.43 (C37), 149.20 (C39), 145.74

(C41), 144.51 (C12), 130.50 (C45), 130.21 (C44), 130.01 (C43), 122.50 (C42), 121.72 (C13),

107.50 (C46), 103.43 (C38), 78.96 (C2), 58.44 (C34), 55.13 (35). MS (ESI+) 660.41 calculated

+ for C41H57ClN2O3 [M+H] , found 660.38.‖

4.12.2. Synthesis of compound VK6

O HO NH Cl OH Cl H O O N N O HO O N DMSO, DCC,DMAP, reflux 120 0C 24 h HO Scheme 4. 14: Synthesis of compound VK6

30 23 47 Cl ―Oleanolic acid (400 mg, 0.8 mmol) 21 22 20 45 46 44 and AEE.Q (220.43 mg, 0.8 mmol) 13 18 19 H 14 12 17 O 49 48 34 N 42 43 25 24 27 33 50 O 35 36 37 41 6 10 11 16 were dissolved in 5 mL of DMSO. 1 5 9 15 O 38 N 31 39 40 2 4 8 29 VK6 DMAP) (97.74 mg, 0.8 mmol) and HO 3 7 32 28 26 DCC (181.56 mg, 0.88 mmol) portions were added and stirred at room temperature for 10 minutes then heated to reflux at 120

0C for 24 hrs, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with DCM (20 mL) and the mixture was washed with 30

92 mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulphate, filtered and concentrated on the rotary evaporator to obtain off-white crystals. The crude compound was purified by column chromatography using silica gel (Merck, silica gel 60 F254: 0.063-0.200 mm). The column was eluted with EtOAc:

0 MeOH (9:1) to afford an off white powder (Yield 51%, m.p 132–133 C, rf value 0.45). IR vmax

3535 (NH), 3391 (OH), 2932-2855 (CH aliphatic), 1704 (C=O), 1626 (C=C alkene), 1451 (C=C

1 aromatic), 1274 (C-N), 628 (C-Cl). H NMR (400 MHz, CDCl3): δ (ppm) 7.10-7.08 (d, J=8.0

Hz, 1H, H-39), 6.85-6.83 (d, J = 8.0 Hz, 1H, H-43), 6.80 (s, 1H, H-46), 6.51-6.50 (d, J = 8.0 Hz,

1H, 44-H), 5.72-5.70 (d, J=8.0, 1H, H-38), 5.37 (t, J = 4.0 Hz, 1H, H-13), 4.16 (q, 4H, H-50),

3.19 (s, 1H, H-36), 3.74 (q, 4H, H-49-34), 3.48 (dd, J=4.0,8.0 Hz, 2H, H-35), (dd, J = 12.0, 4.0

13 Hz, 1H, 18-H), 2.54 (dd, J=4.0,12 Hz, 1H, H-18). C NMR (400 MHz, CDCl3): δ (ppm) 176.99

(C27), 152.48 (C37), 150.11 (C39), 145.13 (C41), 144.41 (C12), 130.21 (C43), 122.48 (C42),

121.99 (C13), 118.46 (C46), 113.43 (C45), 112.48 (C44), 78.96 (C2), 70.23 (C34), 68.11(C49),

+ + 65.13 (C50). MS (ESI ) 704.43 calculated for C43H61ClN2O4 [M+H] , found 704.11.‖

93

4.12.3. Synthesis of compound VK7

O O HO OH OH O O O O O O OH HO O DMSO, DCC,DMAP, reflux 120 0C 24 h HO O O Scheme 4. 15: Synthesis of compound VK7

30 23 ―Oleanolic acid (200 mg, 0.4 21 22 20 59 56 O 58 O55 13 18 19 mmol) and Curcumin (162.09 14 12 17 O 51 36 OH 25 24 27 32 52 50 37 35 57 6 10 11 16 53 47 45 43 41 38 34 mg, 0.44 mmol) was dissolved 1 5 9 15 O 54 31 46 44 42 40 39 2 4 8 29 3 7 O O HO 49 33 48 in 5 mL of DMSO. DMAP) 28 26 VK7 (48.87 mg, 0.4 mmol) and DCC (90.78 mg, 0.4 mmol) portions were added and stirred at room temperature for 10 minutes then heated to reflux at 120 0C for 24 hrs, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with

DCM (20 mL) and the mixture was washed with 30 mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulphate, filtered and concentrated on the rotary evaporator to obtain a light brown powder. The crude compound was purified by column chromatography using silica gel (Merck, silica gel 60 F254:

0.063-0.200 mm). The column was eluted with EtOAc:MeOH (9:1) to afford a pale yellow

0 powder (Yield 53%, m.p 132–133 C, rf value 0.62). IR vmax 3354 (OH), 2929-2898 (CH

1 aliphatic), 1735 (C=O), 1469 (C=C), 1156- 1031(C-O ester). H NMR (400 MHz, CDCl3): δ

(ppm) 7.34-7.32 (d, J=8.0 Hz, 1H, H-46), 6.82-6.80 (d, J = 8.0 Hz, 1H, H-53), 6.78 (s, 1H, H-

50), 6.72-6.70 (d, J=8.40 Hz, 1H, H-54), 6.65-6.63(d, J=8.0 Hz, 1H, H-46), 5.24(t, J=4.0 Hz,

94

1H, H-13), 4.99(s. 1H, H-57), 4.65(s, 1H, H-43), 3.45(s, 6H, H-56 & 59), 3.16-3.12(dd, J=4.0,

13 1H, H-2), 2.83-2.79 (dd, J=4.0 Hz, 1H, H-18). C NMR (400 MHz, CDCl3): δ (ppm) 196.14

(C44), 196.13 (C42), 177.63 (C27), 158.34 (C51), 152.02 (C36), 143.58 (C12), 142.25 (C46),

141.51 (C40), 139.04 (C52), 133.52 (C47), 129.25 (C38), 127.61 (C45), 127.60 (C41),

122.52(C13), 114.13 (C34), 113.24(C37), 113.23(C50), 79.02(C2), 67.96(C59), 58.47(C56). MS

+ + (ESI ) 806.48 calculated for C51H66O8 [M+H] , found 801.14.‖

4.12.4. Synthesis of compound VK8

H OH H O O HO H O H HO 0 DMSO, DCC,DMAP, reflux 120 C 24 h HO Scheme 4. 16: Synthesis of compound VK8

30 23 ―Oleanolic acid (200 mg, 21 58 22 20 47 59 56 48 46 57 13 18 19 32 60 54 0.4 mmol) and ergocalciferol 52 55 14 12 17 O 34 41 43 45 53 44 25 24 27 35 39 42 51 63 6 10 11 16 H 38 49 50 H 1 5 9 15 O 36 61 62 (162.09 mg, 0.44 mmol) 31 37 42 2 4 8 29 VK8 HO 3 7 33 were dissolved in 5 mL of 28 26 DCM. DMAP) (48.87 mg, 0.4 mmol) and DCC (90.78 mg, 0.4 mmol) portions were added and stirred at room temperature for 10 minutes then heated to reflux at 70 0C for 24 hrs, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with DCM (20 mL x 3) and the mixture was washed with 30 mL of ice water to

95 afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulfate, filtered and concentrated on the rotary evaporator to obtain white crystals. The crude was purified by column chromatography using silica gel (Merck, silica gel 60

F254: 0.063-0.200 mm). The column was eluted with Hexane: EtOAc (7:3) to afford white

0 crystals (Yield 77%, m.p 120–122 C, rf value 0.64). IR vmax 3553-3462 (OH), 2930-2854 (CH aliphatic), 1709 (C=O), 1626 (C=C alkene) 1488(C=C aromatic), 1214(C-O ester). 1H NMR

(400 MHz, CDCl3): δ (ppm) 6.22-6.20 (d, J=8.0 Hz, 1H, H-41), 5.86-5.84 (d, J= 8.0 Hz, 1H, H-

42), 5.44-5.42 (d, J=8.0 Hz, 1H, H-53), 5.24 (t, J=4.0 Hz, 1H, H-13), 4.94 (s, 2H, H-40), 4.32

(m, 1H, H-35), 3.16-3.12 (dd, J=4.0 Hz. 1H, H-2), 2.78-2.73 (dd, J=4.0 Hz, 1H, H-18), 2.19(s,

13 1H, H-33). C NMR (400 MHz, CDCl3): δ (ppm) 172.92 (C27), 145.12 (C38), 144.20 (C43),

143.92 (C12), 141.17 (C39), 134.99 (C54), 130.25 (C53), 126.42 (C41), 124.05 (C42), 123.37

+ (C13), 111.12 (C40), 79.02 (C2), 74.75 (C35). MS (ESI ) 834.69 calculated for C58H90O3

[M+H]+, found 834.69.‖

96

4.12.5. Synthesis of compound VK9

HO O C O

O O N OH C HO HN HO N O H Cl N HO HO DMSO, DCC,DMAP, reflux 120 0C 24 h Cl

Scheme 4. 17: Synthesis of compound VK9

29 23 ―Oleanolic acid (200 mg, 0.4 mmol) and 21 22 20 HO O 52 53 51 13 18 19 4-(7-chloroquinolin-4-ylamino)-2- 14 12 17 O 47 24 54 26 50 46 48 6 10 11 16 hydroxybenzoic acid (162.09 mg, 0.44 1 5 9 15 O 45 49 30 44 2 4 8 26 mmol) were dissolved in 5 mL of DMSO. HO 3 7 HN 43 40 31 41 39 27 25 VK9 36 N DMAP) (48.87 mg, 0.4 mmol) and DCC 38 37 35

32 34 (90.78 mg, 0.4 mmol) portions were added 33

Cl 42 and stirred at room temperature for 10 minutes then heated to reflux at 120 0C for 24 hrs, the reaction was monitored by TLC to ensure the completion of reaction, after which the obtained solution was diluted with DCM (20 mL) and the mixture was washed with 30 mL of ice water to afford an organic and aqueous layer. The organic layer was then collected and dried over anhydrous sodium sulfate, filtered and concentrated on the rotary evaporator to obtain a light brown powder. The crude compound was purified by column chromatography using silica gel (Merck, silica gel 60 F254: 0.063-0.200 mm).

The column was eluted with Hexane: EtOAc (7:3) to afford a white powder (Yield 57%, m.p

0 134–135 C, rf value 0.25). IR vmax 3535 (NH), 3391 (OH), 2932-2855 (CH aliphatic), 1704

97

(C=O), 1626 (C=C alkene), 1451 (C=C aromatic), 1274 (C-N), 628 (C-Cl). 1H NMR (400 MHz,

CDCl3): δ (ppm) 7.96-7.94 (d, J=8.0 Hz, 1H, H-39), 7.80 (s, 1H, H-34), 7.56-7.54 (d, J=8.40 Hz,

1H, H-48), 7.43-7.41 (d, J=8.0, 1H, H-37), 7.34- 7.32 (d, J=8.0 Hz, H-32), 5.74 (s, 1H, H-45),

5.64-5.63 (d, J=4.0 Hz, 1H, H- 40 & 49), 5.28 (t, J=4.0 Hz, 1H, H-13), 3.40 (s, 1H, H-43), 3.17-

3.13 (dd, J=4.0 Hz, 1H, H-13), 2.48-2.44(dd, J=4.0 Hz, 1H, H-18), 2.10 (s, 1H, H-31). 13C NMR

(400 MHz, CDCl3): δ (ppm) 176.34 (C26), 171.44 (C51), 157.22 (C46), 151.74 (C39), 149.21

(C41), 148.99 (C44), 148.58 (C35), 144.76 (C12), 136.42 (C33), 128.61 (C48), 127.42 (C34),

122.50 (C13), 118.40 (C37), 112.72 (C40), 112.71 (C49), 111.23 (C47), 105.40 (C45), 78.96

+ + (C2). MS (ESI ) 752.4 calculated for C46H57ClN2O5 [M+H] , found 755.08.‖

98

Unsuccessful hybrid compounds

O

HN

O N H HO HO O OH H H H N O N O O O H2N H O N H HO HSU, DCC, ref 120oC X HO

H H N OH H OH 2 NH O OH H O H HO HSU, DCC, ref 120oC X HO OH OH

N H N NH H HN 2 H OH N O N O H H O O H HO HSU,X DCC, ref 120oC HO

NH2

OH H O H OH O P P OH OH NH HO OH O O H H o OH HO HSU, DCC, ref 120 C HO O O P P OH X OH HO OH

Scheme 4.19: unsuccessful hybrid compounds

99

4.12.6. Antibacterial assay

―The synthesized hybrid compounds were tested against 11 reference bacterial strains namely:

Gram-positive bacteria: Bacillus subtilis (ATCC19659), Enterococcus faecalis (ATCC13047),

Staphylococcus epidermidis (ATCC14990), Staphylococcus aureous, and Mycobaterium smegmatis (MC2155). Gram-negative strains: Enterobacter cloacae (ATCC13047), Proteus vulgaris (ATCC6380), Klebsiella oxytoca (ATCC8724), Proteus vulgaris (ATCC6380),

Pseudomonas aeruginosa (ATCC27853), Proteus mirabilis (ATCC7002) and Escherichia coli

(ATCC25922).‖

4.12.7 Minimum inhibitory concentration (MIC)

―The minimum inhibitory concentration (MIC) of the synthesized hybrid compounds MIC of the tested compounds was carried out following Fonkui et al., (2018). Stock solutions were prepared by adding 4 mL of DMSO to each tube containing in average 20 mg of the synthesized compounds. These solutions were then serially diluted in 100 uL of nutrient broth in a 96 well plates to the desired concentrations (2.5, 1.25, 0.625, 0.3125, 0.1562, 0.0781 mg/mL). Then after, 100 µL of each of these solutions was placed in duplicate and seeded with 100 µL of an overnight bacterial culture brought to 0.5 Mc Farland in nutrient broth. Streptomycin and nalidixic acid were used as positive control and the negative control was prepared to contain

50% nutrient broth in DMSO.

100

CHAPTER FIVE RESULTS AND DISCUSION

5.1. Structural elucidation of compound VK1

39 N 35 Cl 30 23 40 36 34 43 21 22 20 41 37 33 42 38 13 18 19 H 14 12 17 N 46 NH 25 24 27 48 47 45 44 6 10 11 16 1 5 9 15 O31 2 4 8 29 HO 3 7 32 28 26

Figure 5. 1: structural elucidation of compound VK1

The ―IR spectroscopic skeletal vibrations were first used to confirm the important functional groups of compound VK1 (Figure 5.2): 3296 cm-1 (NH stretch) was assigned to the aliphatic

-1 -1 -1 amine; 3076 cm OH stretch; 2932-2873 cm (CH2-CH3) aliphatic stretch; 1626 cm C=O stretch; and peak at 1545 cm-1 frequency further confirmed the C=C aromaticity.

In deuterated chloroform, the structural elucidation of compound VK1 by 1H NMR spectroscopy

(Figure 5.3) showed the expected signals for 4-aminoquinoline aromatic rings in the region

8.28-700 ppm, the olefinic proton of oleanolic acid at 5.28 ppm and hydroxyl proton at 3.16 ppm of oleanolic acid. The doublet at 8.28 ppm with coupling constant J=8.0 Hz and the singlet at

7.32 ppm were assigned to the two protons H-40 and H-48 experiencing the inductive deshielding effect from the ring nitrogen, N-39 and the substituted N-48 respectively. The doublets at 7.18-7.16, 7.08-7.06 and 5.66-5.64 ppm were assigned to the protons H-38, H-33, and H-41 of the aromatic rings respectively. The triplet at 5.28 ppm and two doublets of a

101 doublet at 3.17-3.13 and 2.48-2.43 ppm were assigned to the proton H-13, H-45 and H-18 of oleanolic acid. The multiplet at 4.06-4.00 ppm was assigned to both protons H-2 of oleanolic acid and H-47 of the installed amine group. A singlet at 3.39 ppm was also assigned to the H-48 of the installed amine group.

13C NMR spectrometry was also used to substantiate the structural elucidation of VK1 (Figure

5.4). To the oleanolic acid 13C NMR spectrum; additional aromatic carbon signals were observed. The aromatic signals at 155.23, 152.43, 148.21, 135.43, 129.11, 125.77, 122.53, 119.4, and 114.24 were assigned to the carbons C42, C40, C36, C34, C35, C33, C38, C37 and C41 respectively.

In addition to the IR and NMR spectroscopy experiments, the LC-MS was used to confirm the expected isotopic mass of compound VK1.‖ The determined experimental mass of VK1 [M+H]+

= 673.33 (Figure 5.5) and ―was found to be in good agreement with the theoretical 673.44.‖

100 95 90

85 80 75 70

65 trasmittance% 60 55 50 4000 3500 3000 2500 2000 1500 1000 500 wavenumber cm -1

Figure 5. 2: 1IR spectrum of compound VK1.

102

Figure 5. 3: 1H-NMR spectrum of compound VK1

Figure 5. 4: 13C-NMR spectrum of compound VK1

103

Figure 5. 5: LC-MS results of compound VK1

5.2. Structural elucidation of compound VK2

38 N 34 Cl 23 29 39 35 33 42 21 22 20 40 36 32 41 37 13 18 19 H 14 12 17 N NH 24 26 44 43 6 10 11 16 1 5 9 15 O 2 4 8 28 HO 3 7 31 27 25

Figure 5. 6: structural elucidation of compound VK2

The ―IR spectrum of compound VK2 (Figure 5.7) displayed almost similar peaks with compound VK1, compound VK2 displayed the absorption bands for NH stretch at 3548 cm-1,

OH at 3383 cm-1, CH aliphatic stretch at 2929-2857 cm-1, C=O stretch at 1709 cm-1, C=C aromatic stretch 1631 cm-1 at 1455 cm-1, C-N stretch at 1217 cm-1, and C-Cl stretch at 628 cm-1 which clearly confirms the successful synthesis of compound VK28,13.

104

The 1H NMR and 13C NMR spectra (Figure 5.8 and Figure 5.9) prove that the target compound

1 was achieved. H NMR (400 MHz, CDCl3) spectrum of compound VK2 indicated three doublets at 8.06-8.04 ppm, 7.62-760 ppm and 7.44-7.42 ppm assigned to protons H-39, H-37 and H-32 of the aromatic ring. A singlet at 7.89 ppm belongs to proton H-34. It also indicated another doublet at 5.65-5.63 ppm linked to proton H-40. The triplet at 5.28 ppm is allocated to proton H-13. The two singlets at 3.40 ppm and 2.10 ppm are assigned to proton H-44 of the linker and H-31 hydroxyl of oleanolic acid respectively. Two doublets of doublet at 3.27-3.13 and 2.48-2.43 ppm are linked to protons H-2 and H-18.

13 C NMR (400 MHz, CDCl3) spectrum of compound VK2 (Figure 5.9) showed a signal at

181.52 ppm belonging to the carbonyl carbon (C26). The signals at 152.12 ppm, 149.97 ppm,

148.24 ppm, 135.43 ppm, 129.65 ppm, 127.61 ppm, 122.52 ppm, 118.42 ppm, and 113.65 ppm are allocated to aromatic carbons C39, C41, C35, C33, C34, C32, C37, C36, and C40 respectively. The signals at 144.74 ppm and 121.99 ppm arise from the olefinic carbons C12 and

C13 of oleanolic acid. An oxygenated carbon C2 of oleanolic acid is allocated at 78.97 ppm.

The LC-MS analysis confirmed that compound VK2 was successfully 6,13. MS (ESI+) 631.39

+ calculated for C39H54ClN3O2 [M+H] , found 631.35 (Figure 5.10).‖

105

105

100

95

90

85

80 Transitance % 75

70

65

60 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 5. 7: IR spectra of compound VK2.

Figure 5. 8: 1H NMR Spectrum of compound VK2

106

Figure 5. 9: 13C NMR spectrum of compound VK2.

Figure 5. 10: LC-MS results of compound VK2.

107

5.3. Structural elucidation of compound VK3

34 27 25 26 24 44 45 N43 17 22 23 H 18 16 21 N 1 46 40 29 31 4 2 N 41 39 10 14 15 20 H48 5 9 13 19 42 38 O 3 35 37 Cl 6 8 12 33 47 HO 7 11 36 32 30

Figure 5. 11: Structural elucidation of compound VK3

―The IR spectrum of compound VK3 (Figure 5.12) confirmed the successful formation of VK3 by showing the peaks of the expected functional groups at 3546 (NH), 3359 (OH), 2928-2857

(CH aliphatic), 1712 (C=O), 1637 and 1456 (C=C aromatic), 1223(C-N), 626 (C-Cl).

1 H NMR (400 MHz, CDCl3) spectrum of compound VK3 (Figure 5.13) indicated two singlets with one proton each at 4.45 ppm and 7.62 ppm attributed to protons H-4 of the linker, and H-39 of the aromatic ring. Three doublets at 7.56-7.54, 7.41-7.39, and 5.64-5.62 ppm were assigned to aromatic protons H-42, H-37 and H-45. The triplet with one proton at 5.28 ppm is ascribed to olefinic proton H-17 of oleanolic acid. The singlet with one proton at 3.40 ppm is assigned to proton H-48 of the linker. A multiplet with one proton at 4.32-4.30 ppm is attributed to proton H-

2 of the linker. The two doublets of doublet at 3. 17- 3.13 ppm and 2.48-2.44 ppm were assigned to protons H-6 and H-22 of the oleanolic acid. A singlet with one proton at 2.19 ppm is ascribed to hydroxyl proton H-366,14.

108

13 C NMR (400 MHz, CDCl3) spectrum of compound VK3 (Figure 5.14) showed a signal for carbonyl (C31) at 177.03ppm. It also indicated signals at 155.75, 150.23, and 148.23 ppm attributed to aromatic carbons C38, C42 and C40 respectively. The signals at 144.00 ppm and

121.13 ppm are assigned to olefinic carbons C16 and C17 of oleanolic acid. The signals at

133.75, 128.43, 126.41, 122.50, 119.23, and 114.34 ppm are linked to aromatic carbons C46,

C47, C45, C44, C39, and C43 respectively.

Finally, the LC-MS was further used to confirm the mass of the compound KV3. The determined experimental mass of VK8 [M+H]+ = 671.09 (Figure 5.15) and MS (ESI+) 673.44 calculated for

C42H60ClN3O2.‖

105 100

95 90 85 80

75 Transmitttance% 70 65 4000 3500 3000 2500 2000 1500 1000 500

Wavenumber cm-1

Figure 5. 12: IR spectra of compound VK3.

109

Figure 5. 13: 1H NMR spectrum of compound VK3.

Figure 5. 14: 13C NMR spectrum of compound VK3

110

Figure 5. 15:LC-MS results of compound VK3

5.4. Structural elucidation of compound VK4

7 N 3 Cl 48 41 8 4 2 11 39 40 38 9 5 1 10 6 31 36 37 H 32 30 35 N 16 15 14 NH 43 42 45 18 17 N 13 12 24 28 29 34 H 19 23 27 33 O 49 20 22 26 47 HO 21 25 50 46 44

Figure 5. 16: Structural elucidation of compound VK4

―The IR spectrum of compound VK4 (Figure 5.17) confirmed the successful formation of VK4 by showing the peaks of the expected functional groups at 3356 cm-1 (NH), 3265 cm-1 (OH),

2943-2857 cm-1 (CH aliphatic), 1712 cm-1 (C=O), 1637 cm-1(C=C alkene), 1456 cm-1 (C=C aromatic), 1223 cm-1(C-N), 626 cm-1(C-Cl).

111

The 1H NMR and 13C NMR spectra prove that the target compound VK4 was successfully achieved. 1H NMR (400 Hz) spectrum hybrid compound VK4 (Figure 5.18) presented a doublet with one proton at 7.74-7.72 ppm assigned to proton H-8 of the aromatic ring. A singlet with one proton at 7.14 ppm is assigned toaroamtic proton H-3. Three doublets at 7.04-7.02, 6.74-6.72 and

6.42-6.40 ppm are assigned to aromatic protons H-6, H-1, and H-9. A triplet with one proton at is assigned to the olefinic proton of oleanolic acid H-31. A singlet with one proton at 3.40 ppm is assigned to N-H proton H-12. The multiplet with 4 protons at 4.04 ppm is assigned to both protons H-13 and H-17 of the linker DET. A quartet with 4 protons at 3.66 ppm is linked to both protons H-14 and H-16. The two singlets at 4.45 and 1.98 ppm are linked to protons H-18 and H-

50. Lastly, the two doublets of doublet at 3.14 and 2.45 ppm are assigned to both protons H-36 and H-20.

13 C NMR (400 MHz, CDCl3) spectrum of compound VK4 (Figure 5.19 indicated signals

177.07, 154.65, 151.30, 148.44, 144.75, 134.50, 129.13, 127.41, 122.50, 121.99, 119.09, 113.21,

78.96, 48.08, 47.57, 46.81, 46.23 assigned to carbons C45, C10, C8, C4, C30, C2, C3, C1, C6,

+ + C31, C5, C9, C20, C13, C17, C14, C16. MS (ESI ) 702.46 calculated for C43H63ClN3O2 [M+H] , found 701.38 (Figure 5.20).‖

112

95

85

75

65

55 Transmittance%

45

35 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 5. 17: IR spectrum of compound VK4.

Figure 5. 18: 1H NMR spectrum of compound VK4

113

Figure 5. 19: 13C NMR spectrum of compound VK4.

Figure 5. 20: LC-MS results of compound VK4.

114

5.5. Structural elucidation of compound VK5

30 23 21 22 20

13 18 19 14 12 17 O 35 25 24 27 33 34 36NH 6 10 11 16 1 5 9 15 O31 37 46 Cl 38 42 45 47 2 4 8 29 HO 3 7 39 41 44 32 N 43 28 26 40

Figure 5. 21: Structural elucidation of compound VK5.

―The IR spectrum of compound VK5 (Figure 5.22) confirmed the successful formation of VK5 by showing the peaks of the expected functional groups at 3450 cm-1 (NH), 3307 cm-1 (OH),

2943-2897 cm-1 (CH aliphatic), 1694 cm-1 (C=O), 1468 cm-1(C=C alkene), 1398 cm-1 (C=C aromatic), 1274 cm-1(C-N), 660 cm-1 (C-Cl).

1 H NMR (400 MHz, CDCl3) spectrum of compound VK5 (figure 5.23) indicated a doublet with one proton at 7.94-7.92 ppm assigned to aromatic proton H-39. A singlet with one proton at 7.78 ppm is linked toaroamtic proton H-46. Two doublets with one proton each at 7.58-7.56 and 5.64-

5.62 ppm are attributed to aroamtic protons H-44 and H-38 respectively. A triplet at 5.28 ppm with one proton is assigned to olefinic proton H-13 of oleanolic acid. A singlet with one proton at 4.45 ppm is assigned to proton H-36 of the linker. Three doublets of doublet at 4.34-4.29,

4.05-4.00, 3.17-3.13, and 2.48-2.44 ppm are assigned to protons H-34, H-35, H-2 and H-18 respectively.

115

13 C NMR (400 MHz, CDCl3) spectrum of hybrid compound VK5 (figure 5.24) presented signals at 177.05, 152.43, 149.20, 145.74, 144.51, 130.50, 130.21, 130.01, 122.50, 121.72, 107.50,

103.43, 78.96, 58.44, and 55.13 ppm assigned to carbons C27, C37, C39, C41, C12, C45, C44,

C43, C42, C13, C46, C38, C2, C34, and C35 respectively. MS (ESI+) 660.41 calculated for

+ C41H57ClN2O3 [M+H] , found 660.38 (figure 5.25).‖

100

95

90

85 transmitance%

80

75 4000 3500 3000 2500 2000 1500 1000 500 wavenumber cm-1

Figure 5. 22: IR spectrum of compound VK5.

116

Figure 5. 23:1H NMR spectrum of compound VK5

Figure 5. 24: 13C NMR spectrum of compound VK5

117

Figure 5. 25: LC-MS results of compound VK5

6.6. Structural elucidation of compound VK6

30 23 47 Cl 21 22 20 45 46 44 13 18 19 H 14 12 17 O 49 48 34 N 42 43 25 24 27 33 50 O 35 36 37 41 6 10 11 16 1 5 9 15 O 38 N 31 39 40 2 4 8 29 HO 3 7 32 28 26

Figure 5. 26: Structural elucidation of compound VK6 ―The IR spectrum of compound VK6 (figure 5.27) confirmed the successful formation of VK6 by showing the peaks of the expected functional groups at 3535 cm-1 (NH), 3391 cm-1 (OH),

2932-2855 cm-1 (CH aliphatic), 1704 cm-1 (C=O), 1626 cm-1 (C=C alkene), 1451 cm-1 (C=C aromatic), 1274 cm-1 (C-N), 628 cm-1 (C-Cl).

1 H NMR (400 MHz, CDCl3) spectrum of compound VK6 (figure 5.28) indicated two doublets with one proton each at 7.10-7.08 ppm and 6.85-6.83 ppm assigned to aoamitic protons H-39 and

H-43. A singlet at 6.80 ppm attributed to aromatic proton H-46. Other doublets at 6.51-6.50, and

118

5.72-5.70 ppm linked to proton H-44, and H-38. A triplet with one proton at 5.37 ppm is assigned to olefinic proton H-13 of the oleanolic acid. Two quartets at 4.26 and 3.74 ppm are assigned to protons H-50 and H-34 of the linker respectively. A singlet at 3.92 ppm is linked to proton H-36 of the linker. Three doublets of doublet at 3.48, 3.14, and 2.54 ppm linked to proton

H-35, H-49, and H-18.

13 C NMR (400 MHz, CDCl3) spectrum of hybrid compound VK6 (figure 5.29) presented signals at 176.99, 152.48, 150.11, 145.13, 144.41, 130.21, 122.48, 121.99, 118.46, 113.43, 112.48,

78.96, 70.23, 68.11, and 65.13 ppm linked to carbons C27, C37, C39, C41, C12, C43, C42, C13,

+ + C46, C45, C44, C2, C34, C49, and C50. MS (ESI ) 704.43 calculated for C43H61ClN2O4 [M+H] , found 704.11(figure 5.30).‖

100

95

90

85 Transmitance % 80

75 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 5. 27: IR spectrum of compound VK6.

119

Figure 5. 28: 1H NMR spectrum of compound VK6.

Figure 5. 29: 13C NMR spectrum of compound VK6.

120

Figure 5. 30: LC-MS results of compound VK6

5.7. Structural elucidation of compound VK7

30 23 21 22 20 59 56 O 58 O55 13 18 19 14 12 17 O 51 36 OH 25 24 27 32 52 50 37 35 57 6 10 11 16 O 53 47 45 43 41 38 34 1 5 9 15 54 31 46 44 42 40 39 2 4 8 29 3 7 O O HO 49 33 48 28 26

Figure 5. 31: Structural elucidation of compound VK7 ―The IR spectrum of compound VK7 (figure 5.32) confirmed the successful formation of

VK7 by showing the peaks of the expected functional groups at 3354 cm-1 (OH), 2929-2898 cm-1 (CH aliphatic), 1735 cm-1 (C=O), 1469 cm-1 (C=C), 1156- 1031 cm-1 (C-O ester).

1 H NMR (400 MHz, CDCl3) spectrum of hybrid compound VK7 (figure 5.33) indicated two doublets with one proton each at 7.34-7.32 and 6.82-6.80 ppm linked to proton H-46 and aromatic proton H-53 of the substituted curcumin. A singlet with one proton at 6.78 ppm

121 assigned to aromatic proton H-50. Another two doublets at 6.72-6.70 ppm and 6.65-6.63 ppm linked to aromatic protons H-54, and H-53. A triplet with one proton at 5.24 ppm assigned to olefinic proton H-13 of oleanolic acid. Another three singlets attributed to protons H-57 at

4.99 ppm with one proton, H-43 with one proton at 4.65 ppm and H-56 & 59 at 3.45 ppm with 6 protons. Two doublets of doublet at 3.16-3.12, and 2.83-2.79 ppm assigned to proton

H-2 and H-18 of oleanolic acid.

13 C NMR (400 MHz, CDCl3) spectrum of hybrid compound VK7 ( figure 5.34) presented signals at 196.14, 196.13, 196.13, 177.63, 158.34, 152.02, 142.25, 141.51, 139.04, 133.52,

129.25, 127.61, 127.60, 122.52, 114.13, 113.24, 113.23, 79.02, 67.96, and 58.47 ppm assigned to carbons C44, C42, C27, C51, C36, C12, C46, C40, C52, C47, C38, C45, C41,

C13, C34, C37, C50, C2, C59, and C56 respectively 13,15. MS (ESI+) 806.48 calculated for

+ C51H66O8 [M+H] , found 801.14 (figure 5.35).‖

100

95

90

85 Transmitance %

80

75 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1 Figure 5. 32: IR spectrum of Compound VK7.

122

Figure 5. 33: 1H NMR spectrum of compound VK7

Figure 5. 34: 13C NMR spectrum of compound VK7.

123

Figure 5. 35: LC-MS results of compound VK7

5.8. Structural elucidation of compound VK8

30 23

21 58 22 20 47 59 56 48 46 57 13 18 19 60 54 52 55 14 12 17 O 34 41 43 45 53 44 25 24 27 35 39 42 51 63 6 10 11 16 H 38 49 50 H 1 5 9 15 O 36 37 40 2 4 8 29 HO 3 7 28 26

Figure 5. 36: Structural elucidation of compound VK8

The IR spectrum of compound VK8 (figure 5.37) confirmed a successful isolation of VK8 by showing the peaks of the expected functional groups at 3553-3462 cm-1 (OH), 2930-2854 cm-1 (CH aliphatic), 1709 cm-1 (C=O), 1626 cm-1 (C=C alkene) 1488 cm-1 (C=C aromatic), and 1214 cm-1(C-O ester).

1H NMR (400 Hz) spectrum (figure 5.38) presented two doublets at 6.22-6.20 ppm and 5.86-

5.84 ppm assigned to protons H-41 and H-42 respectively. It also indicated a triplet with one proton at 5.24 ppm which is assigned to olefinic proton H-13 of oleanolic acid. A singlet with

124 two protons at 4.94 ppm is assigned to proton H-40. A multiplet with one proton at 4.32 ppm is assigned to proton H-35. Two doublets of a doublet at 3.16-3.12 ppm and 2.78-2.73 ppm were assigned to protons H-2 and H-18 on oleanolic acid. It also indicated a singlet with one proton at 2.19 ppm which is assigned to proton H-33.

13 C NMR (400 MHz, CDCl3) spectrum of hybrid compound VK8 (figure 5.39) presented signals at 172.92, 145.12, 144.20, 143.92, 141.17, 134.99, 130.25, 126.42, 124.05, 123.37,

111.12, 79.02, and 74.75 ppm assigned to the carbons C27, C38, C43, C12, C39, C54, C53,

C41, C42, C13, C40, C2, and C35 respectively. The determined experimental mass of VK8

[M+H]+ = 834.69 (figure 5.40) and was found to be in good agreement with the theoretical

834.69.‖

100

90

80 %Transmitance 70

60 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber (cm-1)

Figure 5. 37: IR spectrum of compound VK8.

125

Figure 5. 38: 1H NMR spectrum of compound VK8

Figure 5. 39: 13C NMR spectrum of compound VK8

126

Figure 5. 40: LC-MS results of compound VK8

5.9. Structural elucidation of compound VK9

29 23 21 22 20 HO O 52 53 51 13 18 19 14 12 17 O 47 24 54 26 50 46 48 6 10 11 16 1 5 9 15 O 45 49 30 44 2 4 8 HO 3 7 HN 43 40 31 41 39 27 25 36 N 38 37 35

32 34 33

Cl 42

Figure 5. 41: structural elucidation of compound VK9

―The IR spectrum of compound VK9 in figure 5.42 confirmed the successful formation of

VK7 by showing the peaks of the expected functional groups at 3535 cm-1 (NH), 3391cm-1

(OH), 2932-2855 cm-1 (CH aliphatic), 1704 cm-1 (C=O), 1626 cm-1 (C=C alkene), 1451 cm-1

(C=C aromatic), 1274 cm-1 (C-N), 628 cm-1 (C-Cl).

127

1 H NMR (400 MHz, CDCl3): spectrum of compound VK9 (figure 5.43) showed a doublet at

7.96-7.94 ppm assigned to aromatic proton H-39. A singlet with one proton at 7.80 ppm linked to aromatic proton H-34. It indicated another two doublets at 7.56-7.54 ppm and 7.43-

7.41 ppm, and 7.34- 7.32 ppm attributed to aromatic protons H-48, H-37 and H-32 respectively. The two singlets at 5.74 ppm and 5.38 ppm assigned to proton H-45 and H-43.

A doublet and triplet at 5.64-5.63 ppm and 5.28 ppm are assigned to protons H-40/49 and H-

13 respectively. The two doublets of doublet and a singlet at 3.17-3.13, 2.48-2.44 and 2.10 ppm are attributed to protons H-2, H-18 and H-31 of oleanolic acid respectively10,14,16.

13 C NMR (400 MHz, CDCl3) spectrum of hybrid compound VK9 (figure 5.44) presented signals at 176.34, 171.44, 157.22, 151.74, 149.21, 148.99, 148.58, 144.76, 136.42, 127.42,

128.61, 122.50, 118.40, 112.72, 112.71, 111.23, 105.40, and 78.96 ppm assigned to carbons

C26, C51, C46, C39, C41, C44, C35, C12, C33, C48, C34, C13, C37, C40, C49, C47, C45,

+ + and C2 respectively. MS (ESI ) 752.4 calculated for C46H57ClN2O5 [M+H] , found 755.08

(figure 5.45).‖

100

95

90

85

80

75 Transmitance % 70

65

60 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber cm-1

Figure 5. 42: IR spectrum of compound VK9.

128

Figure 5. 43: 1H NMR spectrum of compound VK9.

Figure 5. 44: 13C NMR spectrum of compound VK9

129

Figure 5. 45: LC-MS results of compound VK9

The melting points and the percentage yields of the synthesized compounds range from of

114-135℃ and 51-66% respectively. Hao et al 201317 synthesized a number of hybrid compounds containing oleanolic acid and other scaffolds with the melting point ranging from

120-138%.

LC-MS results successfully confirmed the isolation of hybrid compounds by showing the expected molecular weights that correlate well with calculated molecular weight of the synthesized compounds and some of the compounds were visible as isotopes. The differences on expected molecular weights are linked to the impurities or fragmentation during compound separation via column chromatography.

5.10. Minimum inhibitory concentration

The MIC results of the synthesized compounds are presented in Table 5.1 and Figure 5.46 shows the 96 micro-well plates of the synthesized compounds against the 11 bacterial strained mentioned above.‖

130

Table 5. 1: Antibacterial activities of synthesized compounds.

Minimum inhibitory concentration (MIC, mg/mL)

Test Gram-positive Gram-negative compound

BS EF SE SA MS ECL PV KO PA PM EC

VK1 2.5 1.25 1.25 2.5 2.5 2.5 2.5 1.25 2.5 2.5 1.25 VK2 2.5 1.25 1.25 2.5 1.25 2.5 2.5 1.25 2.5 1.25 1.25 VK3 2.5 2.5 2.5 2.5 2.5 1.25 2.5 2.5 2.5 2.5 2.5 VK4 2.5 1.25 1.25 1.25 1.25 1.25 1.25 1.25 2.5 1.25 1.25 VK5 2.5 1.25 1.25 1.25 1.25 1.25 2.5 1.25 2.5 2.5 2.5 VK6 2.5 1.25 2.5 2.5 1.25 2.5 2.5 2.5 2.5 1.25 1.25 VK7 2.5 1.25 1.25 2.5 1.25 2.5 2.5 1.25 2.5 1.25 1.25 VK8 2.5 1.25 1.25 2.5 1.25 1.25 2.5 1.25 1.25 1.25 2.5 VK9 1.25 2.5 2.5 0.078 1.25 1.25 0.078 1.25 1.25 2.5 1.25 Oleanolic 2.5 2.5 1.25 2.5 1.25 1.25 2.5 2.5 0.078 1.25 2.5 acid STM 16 128 8 256 4 512 128 16 16 128 64 NLD 16 >512 64 64 512 16 128 8 256 32 512

131

Figure 5. 46: The 96 Well plates (MIC) showing the antibacterial activities against 11 bacterial strains. ―MIC values of the synthesized hybrid compounds against eleven bacterial strains are recorded in Table 5.1. The synthesized compounds exhibited significant antibacterial activity against six bacterial strains such as Enterococcus faecalis (EF), Klebsiella oxytoca (KO),

Escherischia coli (EC), Staphylococcus aurous (SA), Proteus vulgaris (PV) and Bacillus subtilis (BS) with greater activity than did the precursor oleanolic acid.

132

VK1, VK2 and VK7 were more effective against Klebsiella oxytoca (KO), Enterococcus faecalis (EF), and Enterobacter cloacae (EC) with the MIC value of 1.25 mg/mL. VK4 was more effective against Staphylococcus aureous (SA), Proteus vulgaris (PV), Klebsiella oxytoca, Enterococcus faecalis, and Enterobacter cloacae with the MIC value 1.25 mg/mL.

VK5 had higher activity against Enterococcus faecalis, Staphylococcus aurous and

Klebsiella oxytoca with MIC value 1.25 mg/mL. VK6 had higher activity against

Enterococcus faecalis and Enterobacter cloacae with MIC value 1.25 mg/mL. VK7 was effective against EF, KO and EC with MIC value of 1.25 mg/mL. VK8 had higher activity against EF and KO with MIC value 1.25 mg/mL. VK9 was synergistic against Bacillus subtilis (BS), SA, PV, KO and EC with MIC value ranging from 1.25 mg/mL to 0.078 mg/mL.

5.11. Successful hybrid compounds

The chemical structures displaced below represent the final hybrid compounds that were successfully synthesized and evaluated for antibacterial activities.

N Cl N Cl

H H N NH N NH

O O HO VK1 HO VK2

N Cl

H N N H N N NH H N O Cl H HO O VK3 HO VK4

133

Cl O NH H O N O Cl O HO O N VK5 N VK6 HO

HO O C O O O O OH O O HO HN O O HO N VK7 VK9

Cl

O H O H

HO VK8

Figure 5. 47: Successful hybrid compounds tested for antibacterial activity.

134

CHAPTER SIX

CONCLUSION AND RECOMMENDATIONS

6.1. Conclusion

Oleanolic acid possesses various biological properties which include anti-bacteria18, antitumor19 and anticancer20. ―Recently, OA was documented as a promising lead compound for new drug formulation19. Indeed, Shirahata et al. (2012)21 demonstrated the improvement of anti-influenza effect of OA by decorating C28 position carboxylic with glycosyl ester moiety, again Pattnaik et al.(2017)22 also decorated C28 of oleanolic acid and their compounds exhibited highly potent activities against the human breast cancer cell lines

(MCF-7 & MDA-MB-231). The present study corroborates the fact that modification of OA in C28 results in enhancement of biological properties. The primary objectives of this work were successfully achieved. The oleanolic acid precursor was successfully isolated from plant

Syzygium aromaticum. The synthesis of hybrid compounds containing the isolated oleanolic acid and other pharmaceutical scaffolds was successfully accomplished and obtained using different reaction routes as described in the dissertation. The characterization and structural elucidation of the novel compounds was carried out using IR, 1H NMR, and 13C NMR spectroscopic methods supported by LC-MS. The antibacterial activity of the synthesized compounds was lastly investigated on various bacterial strains using Minimum inhibition concentration (MIC) method. The synthesized hybrid compounds were more effective compared to oleanolic acid when tested against 11 bacterial strains on both Gram-positive and Gram-negative bacteria. The synthesized compounds were selective and more active against Enterococcus faecalis (EF), Klebsiella oxytoca (KO), Escherischia coli (EC),

Staphylococcus aureous (SA), Proteus vulgaris (PV) and Bacillus subtilis (BS). The

135 synthesized compounds might be helpful in the future development of oleanolic acid analogs as novel antibacterial agents.

6.2. Recommendations

The antibacterial results were excellent indicating the isolated compounds are potent compounds and it also suggests they would exhibit excellent anticancer activity. All the successful hybrid compounds will be taken for anticancer studies. However, more studies are required in order to understand the mode of action of these hybrid compounds.

The synthetic approach needs to be reviewed for the synthesis of the unsuccessful compounds.

136

6.3. References

1. Nqoro X, Tobeka N, Aderibigbe BA. Quinoline-based hybrid compounds with

antimalarial activity. Molecules. 2017;22:2268. doi:10.3390/molecules22122268

2. Souza MVN De, Pais KC, Kaiser CR, Peralta MA, Ferreira MDL, Lourenço MCS.

Synthesis and in vitro antitubercular activity of a series of quinoline derivatives.

Bioorg Med Chem. 2009;17(4):1474-1480. doi:10.1016/j.bmc.2009.01.013

3. Musiol R, Jampilek J, Buchta V, Silva L, Niedbala H, Podeszwa B, Palka A, Majerz-

Maniecka K, Oleksynd B, Polanski J. Antifungal properties of new series of quinoline

derivatives. Bioorg Med Chem. 2006;14:3592-3598. doi:10.1016/j.bmc.2006.01.016

4. Madrid PB, Wilson N, DeRisi JL, Guy RK. NIH Public Access. NIH Public Access.

2015;85(0 1):1-27. doi:10.1016/j.neuroimage.2013.08.045.The

5. Sunduru N, Sharma M, Srivastava K, Rajakumar S, Puri SK, Saxena JK, Prem M. S.

Chauhan PMS. Synthesis of oxalamide and triazine derivatives as a novel class of

hybrid 4-aminoquinoline with potent antiplasmodial activity. Bioorg Med Chem.

2009;17(17):6451-6462. doi:10.1016/j.bmc.2009.05.075

6. Musonda CC, Gut J, Rosenthal PJ,Yardley V, de Souzad RCC and Chibale K.

Application of multicomponent reactions to antimalarial drug discovery . Part 2 : New

antiplasmodial and antitrypanosomal 4-aminoquinoline c - and d -lactams via a ‗ catch

and release ‘ protocol. Bioorg Med Chem. 2006;14:5605-5615.

doi:10.1016/j.bmc.2006.04.035

7. Sibiya HP, Mabandla M V, Musabayane CT. The effects of transdermally delivered

oleanolic acid on malaria parasites and blood glucose homeostasis in P. berghei-

infected male Sprague-Dawley rats. PLoS One. 2016;11(12):1-18.

137

doi:10.1371/journal.pone.0167132

8. Pretorius SI, Breytenbach WJ, Kock C De, Smith PJ, Da DDN. Synthesis ,

characterization and antimalarial activity of quinoline – pyrimidine hybrids. Bioorg

Med Chem. 2013;21(1):269-277. doi:10.1016/j.bmc.2012.10.019

9. Smit FJ, N'Da DD. Synthesis and in Vitro Antimalarial Activity of Novel Chalcone

Derivatives. PhD Thesis, North-West University (Potchefstroom Campus); 2014.

10. Sharma PK, Kumar S, Kumar P, Kaushik P, Dhirender Kaushik D, Dhingra Y, Aneja

KR. Synthesis and biological evaluation of some pyrazolylpyrazolines as anti-

inflammatory – antimicrobial agents. Eur J Med Chem. 2010;45(6):2650-2655.

doi:10.1016/j.ejmech.2010.01.059

11. Chiyanzu I, Clarkson C, Smith PJ, Lehman J, Gut J, Rosenthal PJ, Chibale K. Design,

synthesis and anti-plasmodial evaluation in vitro of new 4-aminoquinoline isatin

derivatives. Bioorg Med Chem. 2005;13:3249-3261. doi:10.1016/j.bmc.2005.02.037

12. Egan TJ, Egan TJ. Haemozoin ( malaria pigment ): a unique crystalline drug target.

Targets. 2003;2(3):115-124.

13. Rali S, Oyedeji OO, Aremu OO, Oyedeji AO, Nkeh-Chungag BN. Semisynthesis of

derivatives of oleanolic acid from Syzygium aromaticum and their antinociceptive and

anti-inflammatory properties. Mediators Inflamm. 2016;2016:1-9.

doi:10.1155/2016/8401843

14. Xiao S, Wang Q, Si L, Zhou X, Zhang Y, Zhang L, Zhou D. Synthesis and biological

evaluation of novel pentacyclic triterpene a -cyclodextrin conjugates as HCV entry

inhibitors. Eur J Med Chem. 2016;124:1-9. doi:10.1016/j.ejmech.2016.08.020

138

15. Wang S, Peng X, Cui L, Li T, Yu B, Ma G, Ba X. Synthesis of water-soluble curcumin

derivatives and their inhibition on lysozyme amyloid fi brillation. Spectrochim Acta

Part A Mol Biomol Spectrosc. 2018;190:89-95. doi:10.1016/j.saa.2017.09.010

16. Bao Z, Xia H, Gui Y, Liu J. Synthesis of azo derivatives of 4-aminosalicylic acid.

chinese Chem Lett. 2007;18:639-642. doi:10.1016/j.cclet.2007.04.031

17. Hao J, Liu J, Wen X, Sun H. Bioorganic & Medicinal Chemistry Letters Synthesis and

cytotoxicity evaluation of oleanolic acid derivatives. Bioorg Med Chem Lett.

2013;23(7):2074-2077. doi:10.1016/j.bmcl.2013.01.129

18. Kurek A, Nadkowska P, Pliszka S, Wolska KI. Modulation of antibiotic resistance in

bacterial pathogens by oleanolic acid and ursolic acid. Phytomedicine. 2012;19(6):515-

519. doi:10.1016/j.phymed.2011.12.009

19. Zhang L, Chen Y, Shi R, Kanga T, Pang G, Wang B, Zhao Y, Zeng X, Zoua C, Wua

P, Li J. Synthesis of hollow nanocages MOF-5 as drug delivery vehicle to solve the

load-bearing problem of insoluble antitumor drug oleanolic acid ( OA ). Inorg Chem

Commun. 2018;96(April):20-23. doi:10.1016/j.inoche.2018.07.029

20. Wiemann J, Heller L, Csuk R. Targeting cancer cells with oleanolic and ursolic acid

derived hydroxamates. Bioorg Med Chem Lett. 2016;26(3):907-909.

doi:10.1016/j.bmcl.2015.12.064

21. Shirahata T, Nagai T, Hirata N, Yokoyama M, Katsumi T. Syntheses and mucosal

adjuvant activity of simplified oleanolic acid possessing cinnamoyl ester.

Bioorg Med Chem. 2017;25(6):1747-1755. doi:10.1016/j.bmc.2016.09.052

22. Pattnaik B, Lakshmi JK, Kavitha R, Bhattacharjee D, Jain N, Uppuluri V. Synthesis ,

structural studies , and cytotoxic evaluation of novel ursolic acid hybrids with

139 capabilities to arrest breast cancer cells in mitosis. J Asian Nat Prod Res.

2017;19(3):260-271. doi:10.1080/10286020.2016.1240169

140

APPENDIX ONE

SPECTRA OF EUGENOL 100

90 3536 3012 2856

3084 1610

2926 80

70 Transmitance %

60 1034 1513 1267

50 4000 3500 3000 2500 2000 1500 1000 500 wavenumber cm--1

Figure 3.9: FTIR spectrum of eugenol.

141

Figure 3. 10: 1H NMR spectrum of eugenol

142

Figure 3.11: 13C NMR spectrum of eugenol.

143

Figure 3.12: : LC-MS results of eugenol.

144

APPENDIX TWO

Spectra of oleanolic acid 100

95

3443 90

85 1466 transmitance% 2870 998

1031 80 1693 2941 75 4000 3500 3000 2500 2000 1500 1000 500 wavenumber/mc-1

Figure 3.13: FT-IR of oleanolic acid.

145

Figure 3.14: 1H NMR spetrum of oleanolic acid.

146

Figure 3.15: 13C NMR spectrum of oleanolic acid

147

Figure 3.16: : LC-MS results of eugenol.

148

APPENDIX THREE

Spectra of Maslinic acid 100

95

3411 90

2884 1464 85 Transmittance%

1279 1053 2940 80

1691

75 4000 3500 3000 2500 2000 1500 1000 500 Wavenumber mc-1

Figure 3.17: FT-IR Spectrum of Maslinic acid.

149

Figure 3.18: 1H NMR Spectrum of Maslinic acid

150

Figure 3.19: 13C NMR Spectrum of Maslinic acid

151

Figure 3.20: 13C NMR Spectrum of Maslinic acid

152

153