Republic of Iraq Kurdistan Regional Government Ministry of Higher Education and Scientific Research University of Sulaimani Faculty of Medical Sciences School of Pharmacy

SYNTHESIS AND CHARACTERIZATION OF NEW COUMARIN DERIVATIVES CONTAINING VARIOUS MOIETIES WITH ANTI BACTERIAL ACTIVITIES

A THESIS SUBMITTED TO THE COUNCIL OF THE SCHOOL OF PHARMACY AT UNIVERSITY OF SULAIMANI IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF MASTER OF SCIENCE IN PHARMACEUTICAL CHEMISTRY

By Shokhan Jamal Hamid B.Sc. Pharmacy 2010

Supervised by Dr. Ammar A. Mahmood Kubba PhD Pharmaceutical Chemistry

Acknowledgements First and foremost, thanks to Almighty God for giving me countless blessing and strength to carry out this study. I would like to express my special appreciation and thanks to my supervisor Dr. Ammar A. Kubba for his guidance, support and patience throughout the study. I owe my sincere and heartfelt thanks to my beloved husband Safeen for his encouragement and countless help in every moment of my study. My great appreciation to Dr. Mohammed Nawzad Sabir, the head of department of Pharmacognosy & Pharmaceutical Chemistry for his tremendous help, support and encouragement. Words fail to express my humble gratitude to Dr. Emad Manhal Al-Khafaji in Faculty of Science/ School of Chemistry for his unlimited help and guidance during my study. I am deeply grateful to public health laboratory of Sulaimani health directorate for allowing me to conduct some works in their laboratories. Many thanks to my friends Swara J. Mohammed and Lana S. Sabir for their help during my study. Last but not least, I would like to thank my family and dear friends. Without their love and affection it would not have been possible to complete this project.

Shokhan J. Hamid

I

Abstract Coumarin is one of the six membered oxygen containing heterocyclic compound and a member of benzo pyrone family. Because of their broad spectrum of biological activities, various coumarin derivatives have been synthesized. In this study some new coumarin derivatives have been synthesized by adding the formyl group to carbon 8 of 7-hydroxy-4-methyl coumarin, using Duff reaction. The series of the reactions for the preparation of new coumarin derivatives was as follows:

Synthesis of 7-hydroxy-4-methyl coumarin [S1] by reaction of resorcinol with ethylaceto acetate, using concentrated sulfuric acid at (0-10) ˚C, then preparation of 8-formyl-7-hydroxy-4methyl coumarin [S2] by the reaction of compound [S1] with hexamine in glacial acetic acid at (85-90) ˚C using 20% hydrochloric acid. Schiff bases [S3-S5] were prepared by the reaction of compound [S2] with different aromatic amines in ethanol without using acid or base, while amino acid Schiff bases [S6-S8] were prepared by the reaction of compound [S2] with different amino acids in basic medium using alcoholic sodium hydroxide solution. The chalcone derivatives [S9-S11] were synthesized by aldol condensation reaction in which compound [S2] was reacted with different aromatic ketones at a temperature not exceeding 25 ˚C using 10% sodium hydroxide solution. Hydrazone derivatives of coumarin [S12-S14] were prepared by refluxing compound [S2] with different hydrazines in absolute ethanol. Compound [S15] which is a hydrazinyl thiazol derivative of coumarin, prepared by the reaction of compound [S14] with chloroacetic acid in glacialacetic acid as solvent containing anhydrous sodium acetate. The progress of the reactions and purity of the synthesized compounds were checked by thin layer chromatography. Structure elucidation was confirmed by physical properties and spectrometric analysis using FT-IR, 13C-NMR and Mass spectroscopy. The new coumarin derivatives have been screened for their antibacterial activity by serial broth dilution method against two gram positive bacteria Staphylococcus epidermidis and Staphylococcus hemolyticus and two gram negative bacteria Escherichia coli and Klebsiella II

pneumoniae. All the synthesized compounds have been found to exhibit considerable antibacterial activity in vitro. Among all the derivatives, compound [S9] showed the highest rate of inhibition against Escherichia coli, while compound [S12] showed greatest anti-bacterial activity against Staphylococcus hemolyticus.

III

Table of Contents Subjects

Page

Acknowledgements

I

Abstract

II

Table of Contents

IV

List of Tables

VII

List of Figures

IX

List of Schemes

XII

List of Abbreviations

XIV

Chapter One: Literature Review 1.1: Heterocyclic Compounds

1

1.2: Coumarin

2

1.3: Structure of Coumarin

2

1.4: Occurrence

3

1.5: Synthesis of Coumarin

5

1.5.1: Perkin Reaction

5

1.5.2: Pechmann Reaction

6

1.5.3: Knoevenagel Condensation

7

1.5.4: Wittig Reaction

8

1.5.5: Kostanecki-Robinson Reaction

8

1.5.6: Reformatsky Reaction

9

1.6: Reactions of Coumarin

10

1.6.1: Reaction with Electrophilic Reagents

10

1.6.2: Reaction with Nucleophilic Reagents

11

1.6.3: Photodimerization of Coumarin

11

1.6.4: Reaction with Dienes (Diels-Alder Reaction)

12

1.6.5: Mannich Reaction

13

1.7: Pharmacokinetics of Coumarin

13

1.7.1: Absorption and Distribution

14

IV

1.7.2: Metabolism

14

1.7.3: Excretion of Coumarin

16

1.8: Toxicities of Coumarin

16

1.9: Biological Activity of Simple Coumarins and Analogues

17

1.9.1: Antimicrobial

18

1.9.2: Antiviral

19

1.9.3: Anticancer

19

1.9.4: Enzyme Inhibition

21

1.9.5: Antioxidant

21

1.9.6: Anti-Inflammatory

22

1.9.7: Anticoagulant

22

1.9.8: Antihyperlipidemic Activity

23

1.9.9: Central Nervous System

24

1.10: Aim of the Study

25

Chapter Two: Experimental 2.1: Chemicals

26

2.2: Instruments and Analytical Techniques

29

2.2.1: Melting Point Apparatus

29

2.2.2: Thin Layer Chromatography (TLC)

29

2.2.3: Infrared Spectrometer

29

2.2.4: 13C-NMR Spectrometer

29

2.2.5: Mass Spectrometer

29

2.3: Procedures

30

2.3.1: Synthesis of 7-hydroxy-4-methyl Coumarin [S1]

30

2.3.2: Synthesis of 8-formyl-7-hydroxy-4-methyl Coumarin [S2]

31

2.3.3: Synthesis of Aromatic Amine Schiff Bases [S3-S5]

32

2.3.4: Synthesis of Amino Acid Schiff Bases [S6-S8]

33

2.3.5: Synthesis of Chalcones [S9-S11]

34

2.3.6: Synthesis of 8-(hydrazonomethyl)-7-hydroxy-4-methyl Coumarin [S12]

35

2.3.7: Synthesis of 8-((2-(2,4-dinitrophenyl)hydrazono)methyl)-7-hydroxy-4-methyl V

Coumarin [S13]

36

2.3.8: Synthesis of 2-((7-hydroxy-4-methyl-2-oxo-2H-chromen-8yl)methylene)

37

hydrazine carbothioamide [S14] 2.3.9: Synthesis of 2-(2-((7-hydroxy-4-methyl-2-oxo-2H-chromen-8-yl)methylene)

38

hydrazinyl)thiazol-4(5H)-one [S15]

Chapter Three: Results and Discussion Results and Discussion

39

3.1: Synthesis of 7-hydroxy-4-methyl Coumarin [S1]

42

3.2: Synthesis of 8-formyl-7-hydroxy-4-methyl Coumarin [S2]

43

3.3: Synthesis of Aromatic Amine Schiff Bases [S3-S5]

48

3.4: Synthesis of Amino Acid Schiff Bases [S6-S8]

55

3.5: Synthesis of Chalcones [S9-S11]

62

3.6: Synthesis of Hydrazone Derivatives [S12-S14]

70

3.7: Synthesis of 2-(2-((7-hydroxy-4- methyl-2-oxo-2H-chromen-8-yl)methylene)

77

hydrazinyl)thiazol-4(5H)-one [S15] 3.8: Anti-microbial Activity

81

3.8.1: Antibacterial Susceptibility Tests

81

3.8.1.1: Dilution methods

81

3.8.1.2: Disk Diffusion Method

82

Chapter Four: Conclusions and Recommendations 4.1: Conclusions

108

4.2: Recommendations

108

References References

109

VI

List of Tables Table

Title

Page

1-1

4

2-1

The four main coumarin subtypes, the main structural features and examples of each coumarin subtype Name of chemicals and their suppliers

26

2-2

physical properties of compound [S1]

30

2-3

physical properties of compound [S2]

31

2-4

physical properties of compound [S3-S5]

32

2-5

physical properties of compound [S6-S8]

33

2-6

physical properties of compound [S9-S11]

34

2-7

physical properties of compound [S12]

35

2-8

physical properties of compound [S13]

36

2-9

physical properties of compound [S14]

37

2-10

physical properties of compound [S15]

38

3-1

FT-IR spectral data of compound [S1]

43

3-2

FT-IR spectral data of compound [S2]

46

3-3

13

48

3-4

FT-IR spectral data of compound [S3-S5]

54

3-5

13

55

3-6

FT-IR spectral data of compound [S6-S8]

61

3-7

13

62

3-8

FT-IR spectral data of compound [S9-S11]

69

3-9

13

70

C-NMR and MS spectral data of compound [S2]

C-NMR and MS spectral data of compound [S3-S5]

C-NMR and MS spectral data of compound [S6-S8]

C-NMR and MS spectral data of compound [S9-S11]

VII

3-10

FT-IR spectral data of compound [S12-S14]

75

3-11

13

76

3-12

FT-IR spectral data of compound [S15]

79

3-13

13

81

3-14

Anti-bacterial activity of compound [S2-S15]

83

3-15

Mean values of MIC

85

C-NMR and MS spectral data of compound [S12-S14]

C-NMR and MS spectral data of compound [S15]

VIII

List of Figures Figure

Title

Page

1-1

Examples of heterocyclic compounds

1

1-2

3

1-3

Chemical structures of benzopyrone subclasses, with the basic coumarin structure (benzo- α -pyrone) [A], and flavonoid (benzo- γ -pyrone) structure [B] Chemical structure of Novobiocin and Clorobiocin

19

1-4

Natural coumarin geiparvarin

20

1-5

Natural coumarin osthenol

21

1-6

Structures of dicoumarol and warfarin

23

1-7

A-coumarin-indole derivative, B-coumarin chalcone fibrate

23

1-8

Natural coumarin, fraxetin

24

3-1

Anti-bacterial activity of compound [S9] against Escherichia coli

84

3-2

Anti-bacterial activity of compound [S15] against Klebsiella pneumoniae

85

3-3

Disc diffusion method of anti-bacterial susceptibility test

86

3-4

Anti-bacterial activity of some coumarin derivatives

87

3-5

FT-IR spectrum of compound [S1]

88

3-6

FT-IR spectrum of compound [S2]

88

3-7

FT-IR spectrum of compound [S3]

89

3-8

FT-IR spectrum of compound [S4]

89

3-9

FT-IR spectrum of compound [S5]

90

3-10

FT-IR spectrum of compound [S6]

90

3-11

FT-IR spectrum of compound [S7]

91

3-12

FT-IR spectrum of compound [S8]

91

IX

3-13

FT-IR spectrum of compound [S9]

92

3-14

FT-IR spectrum of compound [S10]

92

3-15

FT-IR spectrum of compound [S11]

93

3-16

FT-IR spectrum of compound [S12]

93

3-17

FT-IR spectrum of compound [S13]

94

3-18

FT-IR spectrum of compound [S14]

94

3-19

FT-IR spectrum of compound [S15]

95

3-20

13

96

3-21

13

96

3-22

13

97

3-23

13

97

3-24

13

98

3-25

13

98

3-26

13

99

3-27

13

99

3-28

13

100

3-29

Mass spectrum of compound [S2]

100

3-30

Mass spectrum of compound [S3]

101

3-31

Mass spectrum of compound [S4]

102

3-32

Mass spectrum of compound [S5]

102

3-33

Mass spectrum of compound [S6]

103

3-34

Mass spectrum of compound [S7]

104

3-35

Mass spectrum of compound [S10]

105

C-NMR spectrum of compound [S2] C-NMR spectrum of compound [S3] C-NMR spectrum of compound [S5] C-NMR spectrum of compound [S6] C-NMR spectrum of compound [S7] C-NMR spectrum of compound [S10] C-NMR spectrum of compound [S11] C-NMR spectrum of compound [S14] C-NMR spectrum of compound [S15]

X

3-36

Mass spectrum of compound [S11]

105

3-37

Mass spectrum of compound [S13]

106

3-38

Mass spectrum of compound [S14]

106

3-39

Mass spectrum of compound [S15]

107

XI

List of Schemes Scheme

Title

Page

1-1

Perkin synthesis of coumarin

5

1-2

Pechmann synthesis of coumarin

6

1-3

Knoevenagel condensation

7

1-4

Knoevenagel synthesis of coumarin

7

1-5

Wittig synthesis of coumarin

8

1-6

Kostanecki-Robinson synthesis of coumarin

9

1-7

Reformatsky synthesis of coumarin

10

1-8

Reaction of coumarin with electrophile

10

1-9

Reaction of coumarin with nucleophile

11

1-10

Reaction of coumarin with alkali

11

1-11

Photodimerization of coumarin

12

1-12

Photodimerization of coumarin-3-carboxylic acid in the solid state

12

1-13

Addition of dienes to coumarin

13

1-14

Mannich reaction of coumarin

13

1-15

Metabolism of coumarin

15

3-1

Synthesis of coumarin derivatives [S1 and S2]

39

3-2

Synthesis of coumarin derivatives [S3-S11]

40

3-3

Synthesis of coumarin derivatives [S12-S15]

41

3-4

Synthesis of compound [S1]

42

3-5

Mechanism of synthesis of compound [S1]

42

XII

3-6

Synthesis of compound [S2]

43

3-7

Mechanism of synthesis of compound [S2]

44

3-8

Resonance stability of 7-hydroxy-4-methyl coumarin

45

3-9

Synthesis of compounds [S3-S5]

48

3-10

Mechanism of synthesis of aromatic amine Schiff bases [S3-S5]

49

3-11

Synthesis of compounds [S6-S8]

56

3-12

Mechanism of synthesis of amino acid Schiff bases [S6-S8]

57

3-13

Synthesis of compounds [S9-S11]

63

3-14

Mechanism of synthesis of compounds [S9-S11]

64

3-15

Synthesis of compounds [S12-S14]

71

3-16

Mechanism of synthesis of hydrazone derivatives [S12-S14]

71

3-17

Synthesis of compound [S15]

77

3-18

Mechanism of synthesis of compound [S15]

78

XIII

List of Abbreviations Abbreviations

Meaning

µg

Micro gram

µL

Micro liter

3-D

Type of dopaminergic receptor

5HT1A

5-hydroxy tryptamine receptor

Å

Angstrom

CYPs

Cytochrome P family of metabolizing enzyme

D2A

Type of dopaminergic receptor

D3

Type of dopaminergic receptor

DMSO

Dimethyl sulfoxide

DNA

Deoxyribonucleic acid

EFSA

European Food Safety Authority

FDA

Food and Drug Administration

FT-IR

Fourier transform infrared spectroscopy

FT-NMR

Fourier transform -Nuclear magnetic resonance

G+ve

Gram positive

G-ve

Gram negative

HBV

Hepatitis B virus

HIV

Human immunodeficiency virus

HIV-PR

Human immunodeficiency virus protease

LD50

Lethal dose 50

XIV

LNCaP cells

Androgen-sensitive human prostate adenocarcinoma cells

m/z

Mass to charge ratio

MAO

Monoamine oxidase

MHz

Megahertz

MIC

Minimum inhibitory concentration

MP

Melting point

MS

Mass spectrometry

NIOSH

National Institute for Occupational Safety and Health

NMR

Nuclear magnetic resonance

OSHA

Occupational Safety and Health Administration

PABA

Para amino benzoic acid

ppm (δ)

Part per million

QSAR

Quantitative structure activity relationship

Rf

Retention factor

S1-S15

Codes of the synthesized coumarin derivatives

SAIF

Sophisticated analytical instrument facility

SAR

Structure activity relationship

TDI

Tolerable daily intake

TLC

Thin layer chromatography

XV

Chapter One Literature Review 1.1: Heterocyclic Compounds: A cyclic compound containing all carbon atoms in ring formation is referred to as a carbocyclic compound. If at least one atom other than carbon forms apart of the ring system, it is designated as a heterocyclic compound. That is any of a class of organic compounds whose molecules contain one or more rings of atoms with at least one atom (the heteroatom) being an element other than carbon, most frequently oxygen, nitrogen, or sulfur. Since non-carbons are usually considered to have replaced carbon atoms, they are called heteroatoms. The structures may consist of either aromatic or non-aromatic rings. (Figure1-1) shows some examples of heterocyclic compounds (1, 2).

Figure (1-1) Examples of heterocyclic compounds

These compounds generally consist of small (3- and 4- membered) and common (5 to 7 membered) ring system. The aromatic heterocyclic compounds are those which have a heteroatom in the ring and behave in a manner similar to benzene in some of their properties. Furthermore, these compounds obey with the general rule proposed by Hückel. This rule states that aromaticity is obtained in cyclic conjugated and planar systems containing (4n+2) π electrons. The conjugated cyclic rings contain six π-electrons as in benzene. This forms a conjugated molecular orbital system which is thermodynamically more stable than the non-cyclic conjugated system (3).

1

This extra stabilization results in a diminished tendency of the molecule to react by addition but a large tendency to react by substitution in which the aromatic ring remains intact (4). A heterocyclic ring may comprise of three or more atoms which are saturated or unsaturated. Also, the ring may contain more than one heteroatom which may be similar or dissimilar. The chemistry of heterocyclic compounds is as logical as that of alphabetic or aromatic compounds. A large number of heterocyclic compounds are essential to life. Various compounds such as alkaloids, antibiotics, essential amino acids, vitamins, haemoglobin, hormones and a large number of synthetic drugs and dyes contain heterocyclic ring system. Knowledge of heterocyclic chemistry is useful in biosynthesis and drug metabolism as well (2, 5).

1.2: Coumarin: Coumarin is one of six-membered oxygen containing heterocyclic compounds. It is a fragrant organic chemical compound which is a natural substance found in many plants

(6)

. The name

comes from a French term for tonka bean, coumarou, one of the sources from which coumarin was first isolated as a natural product in 1820 (7).

1.3: Structure of Coumarin: Coumarin is classified as a member of the benzopyrone family of compounds which consist of a benzene ring joined to a pyrone ring. The benzopyrones can be subdivided into the benzo-αpyrones to which the coumarins belong and the benzo-γ-pyrones, of which the flavonoids are principal members (Figure 1-2) (8).

2

Figure (1-2) Chemical structures of benzopyrone subclasses with the basic coumarin structure (benzo- α -pyrone) [A], and flavonoid (benzo- γ -pyrone) structure [B]

1.4: Occurrence: The coumarins can be categorized into four sub-types: the simple coumarins, furanocoumarins, pyranocoumarins and the pyrone-substituted coumarins (Table 1-1). The simple coumarins e.g. coumarin, 7-hydroxycoumarin and 6,7-dihydroxycoumarin, are the hydroxylated, alkoxylated and alkylated derivatives of the parent compound coumarin, along with their glycosides. Furanocoumarins consist of a five-membered furan ring attached to the coumarin nucleus, divided into linear or angular types with substituents at one or both of the remaining benzoid positions. Pyranocoumarin members are analogous to the furanocoumarins, but contain a six-membered ring. Coumarins substituted in the pyrone ring include 4hydroxycoumarin (7, 8).

3

Table (1-1) the four main coumarin subtypes, and the main structural features and examples of each coumarin subtype

Classification

Features

Examples

Simple coumarins

Hydroxylated, alkoxylated or alkylated on benzene ring 7-hydroxycoumarin

Furanocoumarins

5-membered furan ring attached to benzene ring. Linear or angular Psoralen

Pyranocoumarins

Angelicin

6-membered pyran ring attached to benzene ring. Linear or angular Seselin

Pyrone-substituted

Substitution on pyrone ring,

coumarins

often at 3-C or 4-C positions

Xanthyletin

Warfarin

4

1.5: Synthesis of Coumarin: The biosynthesis of

coumarin

and cyclization of cinnamic acid

in

plants

is

via hydroxylation, glycolysis,

(9)

. Coumarins chemically can be classically synthesized by the

Perkin, Pechmann or Knoevenagel reactions. Recently, the Wittig, the Kostanecki–Robinson and Reformatsky (9-11). However the two most important methods for the synthesis of coumarin derivatives are Perkin and Pechmann reactions (11).

1.5.1: Perkin Reaction: In 1868, Perkin for the first time reported the synthesis of coumarin (12). In the Perkin reaction coumarin was synthesized by aldol condensation of aromatic salicylaldehyde and acid anhydrides, in the presence of an alkali salt of the acid such as sodium acetate, as shown in (Scheme 1-1) (13, 14).

Scheme (1-1) Perkin synthesis of coumarin

5

1.5.2: Pechmann Reaction: A very valuable method for the synthesis of coumarins is the Pechmann reaction. In general, the coumarins were obtained by condensation of phenols with β-ketoesters, in the presence of acid catalysts. The reaction is often called Pechmann-Duisberg, when acetoacetic esters and derivatives are used (Scheme 1-2) (9, 10).

The reaction is conducted with a strong Brønstedt acid such as Lewis acid; AlCl3. The acid catalyses transesterification as well as keto-enol tautomerisation:

A Michael Addition leads to the formation of the coumarin skeleton. This addition is followed by rearomatisation:

Subsequent acid-induced elimination of water gives the product:

Scheme (1-2) Pechmann synthesis of coumarin 6

1.5.3: Knoevenagel Condensation: Condensation of aldehydes with active methylene compounds, in the presence of ammonia or amines, is a reaction known as Knoevenagel. Usually, the reaction is catalyzed by weak bases or by suitable combinations of amines and carboxylic or Lewis acids under homogeneous conditions. Moreover, when malonic acid and pyridine, with or without traces of pipiridine, are used the reaction is often named as Doebner modification (Scheme 1-3). A large list of coumarins has been synthesized using this method, for instance, coumarin-3-carboxylic acids, amino- and alkyl amino coumarins and 3-acetylcoumarins (15).

Scheme (1-3) Knoevenagel condensation

In the early 1900s, the Knoevenagel reaction emerged as an important synthetic method to synthesize coumarin derivatives which was substituted at the 3-position (Scheme 1-4) (16, 17).

Scheme (1-4) Knoevenagel synthesis of 3-substituted coumarin 7

1.5.4: Wittig Reaction: In the Wittig reaction, the alkene formation occurs from carbonyl compounds and phosphonium ylides, proceeding primarily through betaine and/or oxaphosphetane intermediates. When the ylide is replaced by a phosphine oxide carbanion or by a phosphonate carbanion, the reaction is referred to Horner or Horner-Emmons- Wadsworth, respectively

(15)

. This type of

olefination of ortho-hydroxy carbonyl aromatic compounds, followed by further lactonization, is a well-known method for the preparation of coumarin derivatives (Scheme 1-5) (17).

Scheme (1-5) Wittig synthesis of coumarin

1.5.5: Kostanecki-Robinson Reaction: The formation of coumarins, usually 3- and 4-substituted coumarins, by this reaction proceeds via acylation of ortho-hydroxy aryl ketones with aliphatic acid anhydrides, followed by cyclization (Scheme 1-6) (15).

8

Scheme (1-6) Kostanecki-Robinson synthesis of coumarin

1.5.6: Reformatsky Reaction: Condensation of aldehydes or ketones with organozinc derivatives of α-halo esters to yield βhydroxy esters is known as the Reformatsky reaction. In appropriate reaction conditions, lactonization could occur with the formation of coumarins (Scheme 1-7) (18).

9

Scheme (1-7) Reformatsky synthesis of coumarin

1.6: Reactions of Coumarin: 1.6.1: Reaction with Electrophilic Reagents: Coumarin and its derivatives undergo reactions at the activated C-3,4 double bond, or they undergo electrophilic aromatic substitution on the benzene ring most preferably at C-6 and C-8 (19)

. Chlorosulphonation with chlorosulphonic acid gives coumarinsulphonyl chloride in good

yield (Scheme 1-8) (20).

Scheme (1-8) Reaction of coumarin with electrophile

10

1.6.2: Reaction with Nucleophilic Reagents: Severel kinds of nucleophile may react with coumarin either at C-2 (a) or C-9 (b). Some of these reactions involve ring opening (Scheme 1-9) (21).

Scheme (1-9) Reaction of coumarin with nucleophile

The substituted coumarin was converted into benzofuran-2-carboxylic acid upon heating with alkali at 100 ˚C as outlined in (Scheme 1-10) (20).

Scheme (1-10) Reaction of coumarin with alkali

1.6.3: Photodimerization of Coumarin: It is usually difficult to control the regio- and stereoselective [2+2] photodimerization of coumarins both in solution and in the solid state (Scheme 1-11). For example, the direct photoirradiation of coumarin in benzene afforded a mixture of syn–head–head, anti–head–head, syn–head–tail and anti–head–tail (22, 23).

11

Scheme (1-11) Photodimerization of coumarin

More recently, when monomer pairs of coumarin-3-carboxylic acid are arranged in crystals such that the C=C double bonds are related by an inversion center and separated by 3.632 Å, the [2+2], cycloaddition reaction was achieved upon irradiation in the solid state (Scheme 1-12) (23).

Scheme (1-12) Photodimerization of coumarin-3-carboxylic acid in the solid state

1.6.4: Reaction with Dienes (Diels-Alder Reaction): Numerous atempts to bring about the addition of dienens to coumarin as [2+4] cycloaddition, which would yield directly hydroginated di-benzopyrons were failed under normal conditions. The reaction was finally accomplished in the case of 2,3-dimethyl-1,3-butadiene, but not with

12

butadiene or isoprene, by using a large excess of reagent and a tempeature of 260 ˚C (Scheme 113) (24).

Scheme (1-13) Addition of dienes to coumarin

1.6.5: Mannich Reaction: Mannich

reported

that

4-hydroxy coumarin

reacts

with

secondary amines

and

paraformaldehyde to form substituted 3-methyl amino coumarin (Scheme 1-14) (25).

Scheme (1-14) Mannich Reaction of coumarin

1.7: Pharmacokinetics of Coumarin: The pharmacokinetics of coumarin, including the excretion of various metabolites, was elucidated over many years. Coumarin is rapidly and almost completely metabolised with little unchanged compound excreted (8).

13

1.7.1: Absorption and Distribution: Following oral administration, coumarin is rapidly absorbed from the gastrointestinal tract and is distributed throughout the body

(26)

. Coumarin and 7-hydroxycoumarin are both poorly

soluble in water 0.22 and 0.031 %, respectively. However, both compounds have high partition coefficients 21.5 for coumarin and 10.4 for 7-hydroxycoumarin, which are considered favorable for the rapid absorption of compounds once they are in solution. This coupled with the fact that coumarin is non-polar. It also suggests that theoretically coumarin should cross lipid bilayers easily by passive diffusion (8, 27). Pharmacokinetic studies in humans have demonstrated that coumarin is completely absorbed from the GIT after oral administration, and extensively metabolised by the liver in the first pass with only between 2-6 %, reaching the systemic circulation intact. The low bioavailability of coumarin, in addition to its short half-life (1.02 hrs. per oral and 0.8 hrs. intravenous), has brought into question its importance in vivo. At normal therapeutic plasma concentrations, many drugs exist in the plasma mainly in the bound form. It has been shown that 35% of coumarin and 47% of 7-hydroxycoumarin bind plasma proteins

(28)

. Availability of the compounds at their

target tissues should not be problematic since the proportions that bound are well below the accepted critical value of 80% binding. The pharmacokinetics of coumarin has been studied in a number of species including the rat, dog, rhesus monkey and in man (27).

1.7.2: Metabolism: Traditionally coumarin has been viewed by pharmacologists as the ideal model for studying the complex metabolism of a structurally simple organic molecule. Therefore, its metabolic fate has been extensively studied. Determining the metabolic fate of coumarin is important in order to utilize the fact that it is metabolized at several sites, and to access the possible dependence of coumarin-induced toxicity on metabolism (29). The superfamily of cytochromes P450 (CYPs) consists of microsomal haemoproteins that catalyse the oxidative, peroxidative and reductive metabolism of a wide variety of endogenous and exogenous compounds. The CYP superfamily is divided into families and subfamilies according to their nucleotide sequence homology. Coumarin is metabolized initially by specific cytochrome P-450-linked mono-oxygenase enzyme CYP2A6 system in liver microsomes resulting in hydroxylation to form 7-hydroxycoumarin. After 7-hydroxylation, coumarin 14

undergoes a phase II conjugation reaction resulting in a glucuronide conjugation associated with 7-hydroxycoumarin. The 7-hydroxylase activity is exceptionally high in human liver microsomes compared with its activity in the livers of other animal species. The activity of coumarin 3hydroxylase is very high in rodent microsomes, but is absent in human microsomes. Although coumarin may be metabolised by hydroxylation at all six possible positions (carbon 3, 4, 5, 6, 7, and 8), the most common routes of hydroxylation are at positions 7 and 3 to yield 7hydroxycoumarin and 3- hydroxycoumarin respectively (Scheme 1-15). 7-hydroxylation has received the most attention among the various metabolic steps. This is predominantly because it is the major metabolic route in humans and is easily analyzed (8, 29).

Scheme (1-15) Metabolism of Coumarin

15

1.7.3: Excretion of Coumarin: According to the studies done on the excretion of coumarin, 7-hydroxycoumarin,and 4hydroxycoumarin in chickens by use of the Sperber technique indicates that the chicken excreted these coumarins almost entirely in conjugated forms (30). Both 7-hydroxycoumarin and 4-hydroxycoumarin appeared in the urine only as the corresponding glucuronide. During infusion, 84% of excreted radioactivity was identified as 7hydroxycoumarin -glucuronide with only 8% appearing as a conjugate of open-ring metabolite ohydroxyphenylacetic acid (30, 31).

1.8: Toxicities of Coumarin: Coumarin is moderately toxic to the liver and kidneys with a median lethal dose (LD50) of 275 mg/kg. Coumarin is hepatotoxic in rats but less so in mice. Rodents metabolize it mostly to 3,4-coumarin epoxide, a toxic and unstable compound that on further differential metabolism may cause liver cancer in rats and lung tumors in mice. Humans metabolize it mainly to 7hydroxycoumarin, a compound of lower toxicity (26). The European Food Safety Authority (EFSA) adopted in 2004 an opinion in which it was concluded that coumarin was not genotoxic in experimental animals and therefore, a tolerable daily intake (TDI) could be allocated (32). The German Federal Institute for Risk Assessment has established a tolerable daily intake (TDI) of 0.1 mg coumarin per kg body weight, but also advises that higher intake for a short time is not dangerous. The Occupational Safety and Health Administration (OSHA) of the United States does not classify coumarin as a carcinogen for humans (6, 33). Coumarin

is

found naturally in

currants, apricots, and cherries

many edible plants

such as strawberries, black

(7)

. Coumarin is often found in artificial vanilla substitutes,

despite having been banned as a food additive in numerous countries since the mid-20th century. In 1954 the United State Food and Drug Administration (FDA) classified coumarin as a toxic substance and banned its use in foods, mainly because of the hepatotoxicity results in rodents. Coumarin is currently listed by the Food and Drug Administration (FDA) of the United States among substances generally prohibited from direct addition or use as human food. This

16

move was not copied by their European counterparts until 20 years later. The FDA action was taken based on results from routine toxicity tests which showed that coumarin initiated toxic liver damage in rats. No pathological effects were observed in any other organs (26). Clinically, studies carried out on humans have shown little evidence of liver dysfunction. An idiosyncratic-type hepatotoxicity was observed in a clinical trial of 2163 patients, but with only a 0.37% incidence. In addition, cessation of coumarin therapy returned the elevated liver enzymes to normal levels in patients with this response (34). Coumarin has been cited as a chemical carcinogen by National Institute for Occupational Safety and Health (NIOSH), and has been reported as a probable carcinogen in rodent carcinogenicity bioassays. However, as precaution needs to be taken in extrapolating this information to human situations as these tests were carried out in rodents. Various tests have shown that coumarin and its metabolites are non-mutagenic. Although coumarin can cause chromosome and DNA breaks in plant cells, it offers anti-mutagenic protection in E. coli cells exposed to U.V. radiation or 4-nitro-quinoline-l-oxide

(35)

. In cultured Chinese hamster ovary

cells, neither coumarin nor 7-hydroxycoumarin induces sister chromatid exchange. Coumarin is also found to be non-teratogenic and non-phototoxic (36).

1.9: Biological Activities of Simple Coumarins and Analogues: The coumarins are of great interests due to their biological properties. In particular, their physiological, bacteriostatic and anti-tumor activities make these compounds attractive for further backbone derivatization and screening as novel therapeutic agents (37). Coumarin and its derivatives have antimicrobial, antiviral, anti-inflammatory, antioxidant, anticoagulant and enzyme inhibition activities

(38-44)

. It has been shown that coumarin and its

metabolite 7-hydroxycoumarin have antitumor activity against several human tumor cell lines. In addition, it has been shown that 4-hydroxycoumarin and 7-hydroxycoumarin inhibited cell proliferation in a gastric carcinoma cell line (44).

17

1.9.1: Antimicrobial: Novobiocin and clorobiocin (Figure 1-3) are coumarin antibiotics of natural origin, which are inhibitors of DNA gyrase, and have a broad spectrum towards Gram-positive bacteria, including methicillin resistant strains of staphylococci species. Due to some limitations of these compounds, particularly with regard to solubility, toxicity and development of resistance, a novel series of coumarin analogues has been synthesized (15). Over the past years, several efforts were directed towards the design of effective, orally bioavailable coumarin antibiotic inhibitors of bacterial DNA gyrase. From these studies, a series of coumarin congeners has appeared that bear the coumarin part of the molecule and isosteres, like carboxyl or basic amino groups, in order to improve the antibacterial activity. Molecular modeling has been used in conjunction with crystallographic and biological data. In an attempt to design compounds that mimic the structure and activity of the coumarin antibiotic novobiocin, possessing a better profile. The novobiocin analogues, bearing different amino groups, displayed excellent inhibitory activity against DNA gyrase supercoiling. The introduction of dialkyl substituents at the 5,5´-position of novobiose leads to coumarin analogues with improved in vitro antibacterial activity incorporation of alkyl, alkylamino and aryl substituents afford new analogues of coumarin containing antibiotics with interesting activities. A series of novobiocinlike coumarin carboxylic acids has also been synthesized bearing L-rhamnosyl moiety, instead of L-novobiose, as the sugar portion of the molecule. It was found that when alkyl side-chains at C5 of coumarin were introduced, an enhancement of the in vitro antibacterial properties was observed. Several coumarin derivatives were also tested for their antifungal activity. A free 6OH on the coumarin nucleus was found to be important for its antifungal activity. Also, a free 7OH on the same nucleus, was important for antibacterial activity (39, 41, 42, 45-49).

18

Figure (1-3) Chemical structure of Novobiocin and Clorobiocin

1.9.2: Antiviral: The antiviral activity of simple coumarins focuses essentially on the inhibition of HIV-1 protease (HIV-PR) and HIV-1 integrase. The recent advances in the development of coumarin derivatives as potent anti-HIV agents concerning the discovery, structural modification and structure-activity relationships studies have been topics of different reviews or updated articles. With this goal, the inhibitory activity of various coumarins towards HIV-1 protease has been investigated and classified as a class of drugs of interest as antiviral agents. From this, phenprocoumon, warfarin and substituted 4-hydroxy-2-pyrone derivatives are actually referred to as first generation of HIV-PR inhibitors. It was also found that certain coumarin dimers, particularly those containing hydrophobic moieties, display potent inhibitory activity against HIV-1 integrase. In addition, some coumarin derivatives were tested for their activity against Herpes simplex virus. From these studies, 5,7,4´-trihydroxy-4-styrylcoumarin was found to exhibit a significant antiviral activity. It is worthwhile to note that the natural collinin; greveal has significant anti-HBV, DNA replication activity (50, 51).

1.9.3: Anticancer: Among the coumarins screened for anticancer activity, geiparvarin, (Figure 1-4) was found to be the most representative. Geiparvarin is a natural coumarin based structure, isolated from the leaves of Geijera parviflora, which is known for its significant in vitro cytostatic activity. The compound is constituted of three units: a furan, an unsaturated alkenyloxy substituent and a 19

coumarin moiety. The first mentioned unit is essential for the activity as it could work as an alkylating agent of bionucleophiles through a Michael-type reaction. In that way, geiparvarin became a challenging lead compound, for the synthesis of analogues that could also be promising candidates for anticancer activity. (52).

Figure (1-4) Natural coumarin, geiparvarin

Moreover, the inhibitory effects of several simple coumarins, synthesized or isolated from plants, on cytotoxic activity have also been reported (44, 53-58). The in vitro effects of 4-hydroxycoumarin were studied, employing melanoma and the nonmalignant fibroblastic cell lines. It was proposed that the compound might be a useful adjuvant for melanoma therapy (59-61). Hydroxycoumarins with a nitro group in the aromatic ring have been shown to be selective anti-proliferative agents that mediate apoptosis in renal carcinoma cells, through modulation of mitogen-activated protein kinases. It is important to note that the chemo preventive effects of several natural coumarins have been evaluated (7). The mutagenicity of simple coumarins has also been studied. It was found that the antimutagenicity of this class of compounds was linked to the presence of polar functions at carbons 3, 4, and 7 (47, 62).

20

1.9.4: Enzyme Inhibition: Some natural and synthetic coumarins have been found to be cholinesterase inhibitors, which are considered to be a promising approach for the treatment of Alzheimer´s disease, and for possible therapeutic applications in the treatment of Parkinson’s disease. Steroid 5α-reductase type I inhibitory activities of a series of umbelliferone derivatives were evaluated in cell culture systems. A SAR study was performed in order to elucidate the essential structural requirements for the activity. A natural prenylated coumarin, osthenol was also revealed to be a new potential inhibitor of 5α-reductase type I in LNCaP cells (63).

Figure (1-5) Natural coumarin, osthenol

1.9.5: Antioxidant: Various synthetic simple coumarins with various hydroxyl groups and other substituents were tested for their antioxidant activity namely, in relation to the ability to inhibit lipid peroxidation and to scavenge reactive species, for instance, hydroxyl and superoxide radicals, and hypochlorous acid (44, 64, 65). Several

coumarins

have

shown

beneficial

biochemical

profiles

in

relation

to

pathophysiological processes dependent upon reactive oxygen species. Classic and three dimensional (3-D) QSAR analyses of radical scavengers, structurally based on coumarin, have also been performed. The photodynamic damage prevention done by some hydroxy coumarins was evaluated, and compared with that of p-aminobenzoic acid (PABA) as a model sun screen. The activity could be related to their antioxidant action which could minimize skin photo aging (66-69)

.

21

1.9.6: Anti-Inflammatory: It has been found that several coumarins isolated from plants or of synthetic origin possess significant anti-inflammatory and/or analgesic activities

(40, 70-72)

. In a QSAR study for lead

optimization in the design of coumarins as potent non-steroidal anti-inflammatory agents, it was found that substituents in the coumarin positions C-4 and C-7 contributed to the high activity. New diamino ether coumarin derivatives with anti-inflammatory and antioxidant activity were also synthesized. Lipophilicity and ionization were found to be important physicochemical parameters which correlated with the biological activity (15).

1.9.7: Anticoagulant: Warfarin (Fig. 1-5) is a synthetic derivative of dicoumarol, a 4-hydroxycoumarin derived mycotoxin anticoagulant originally discovered in spoiled sweet clover-based animal feeds. It was initially introduced in 1948 as a pesticide against rats and mice, and is still used for this purpose. In the early 1950s, warfarin was found to be effective and relatively safe for preventing thrombosis and thromboembolism in many disorders. It was approved for use as a medication in 1954, and has remained popular ever since. It is considered, even today, the dominant coumarin anticoagulant, owing to its excellent potency and good pharmacokinetic profile.

Warfarin

blood coagulation by

and

related 4-hydroxycoumarin-containing

inhibiting vitamin

K

epoxide

reductase.

molecules It

is

decrease

an enzyme that

recycles oxidized vitamin K1 to its reduced form, after it has participated in the carboxylation of several blood coagulation proteins, mainly prothrombin and factor VII (73, 74). In the search for platelet aggregation inhibitors, several coumarins, either of natural or synthetic origin has been tested. A number of coumarin compounds possessing anticoagulant activity, like their prototype dicoumarol (Figure 1-5), have been synthesized as potential drugs for the management of myocardial infarction (26, 70).

22

Figure (1-6) Structures of dicoumarol and warfarin

1.9.8: Antihyperlipidemic Activity: Statins constitute the major therapeutic force for treatment of hyperlipidemia. However, these have some side effects like muscle fatigue, type 2 diabetes, liver damage and digestive problems. Naturally derived coumarins such as esculetin inhibit oxidative low density lipoproteins and umbelliferone possess good lipid lowering potential. Many synthetic derivatives of coumarins have also emerged as good leads (75). Hybridization of coumarin with an indole moiety (Figure 1-6), which is present in various synthetic statins like fluvastatin has yielded coumarin–indole hybrids which at the dose of 10 mg/kg significantly decrease the plasma triglycerides and total cholesterol by 55% and 20%, respectively. Coumarin-chalcone fibrates (Figure 1-6), has also been synthesized which inhibit the biosynthesis of cholesterol and enhances the activity of lipolytic enzyme, lipoprotein lipase, to facilitate early clearance of lipids from circulation in triton induced hyperlipidemia (66).

Figure (1-7) A-coumarin-indole derivative

B-coumarin chalcone fibrate

23

1.9.9: Central Nervous System: Phenylpiperazine coumarin derivatives were synthesized, and the affinities of these compounds for dopaminergic (D2A, D3) and serotonergic (5HT1A) receptors were evaluated. The results obtained confirm the importance of the N-arylpiperazine fragment in the modulation of this type of activity. SAR studies on antipsychotic compounds were also performed, in which several 4-cyclohexyl hydroxy coumarins were engaged. The potential neuro-protective effects of the natural coumarin; fraxetin (Figure 1-7) on the signalling pathways using a neuronal cell model of Parkinson´s disease were also studied (26, 50, 76).

Figure (1-8) Natural coumarin, fraxetin

24

1.10: Aim of the Study: Coumarin and its derivatives are important organic compounds that have significant biological activities. On the bases of earlier reviews that proved this fact, we synthesized some new (Schiff bases, chalcones, hydrazones and cyclized thiosemicarbazone) derivatives at C-8 of coumarin nucleus, and evaluated their anti-bacterial activity by serial broth dilution method.

25

Chapter Two Experimental 2.1: Chemicals: The specific chemicals used in this work are listed in table (2-1) with their chemical structures and suppliers. Table (2-1) Name of chemicals and their suppliers Chemicals

Structure

Supplier

2,4-dinitro phenyl hydrazine

Alpha Chemika/ India

2-hydroxy acetophenone

Sigma-Aldrich

4-nitro aniline

Sigma-Aldrich

7-hydroxy-4-methyl coumarin

Sigma-Aldrich

Absolute ethanol

J.T.Baker/ Netherland

Acetophenone

Sigma-Aldrich

Alanine

Merck

Anhydrous calcium chloride

Ca-Cl2 26

Alpha Chemika/ India

Anhydrous sodium acetate

Scharlau/ Spain

Aniline

AVONCHEM/ UK

Chloroacetic acid

Sigma-Aldrich

Chloroform

Merck

Dehydroacetic acid

Sigma-Aldrich

Diethyl ether

Alpha Chemika/ India

Ethyl acetate

Merck

Ethyl acetoacetate

Merck

Glacial acetic acid

AVONCHEM/ UK

Glycine

Merck

Hexamethylene tetramine

Sigma-Aldrich

(Hexamine)

Hydrazine hydrate 99%

NH2-NH2.H2O

Sigma-Aldrich

Hydrochloric acid

Alpha Chemika/ India

Methanol

Alpha Chemika/ India 27

Para amino benzoic acid

Sigma-Aldrich

Phenyl alanine

Sigma-Aldrich

Resorcinol

Merck

Sodium hydroxide

NaOH

Fluka

Sulphuric acid

Alpha Chemika/ India

Thiosemicarbazide

Sigma-Aldrich

Note: The solvents used during the work were purified by distillation.

28

2.2: Instruments and Analytical Techniques: 2.2.1: Melting Point Apparatus Melting points were determined by using Stuart/ SMP3 melting point apparatus version 5.0 in open capillary tubes, and are uncorrected.

2.2.2: Thin Layer Chromatography (TLC) The purity of the synthesized compounds and progress of the reaction were determined using Thin Layer Chromatography on aluminum silica gel 60 florescent plate254 nm (Fluka) detected by UV light (302 nm).

2.2.3: Infrared Spectrometry Infrared spectra were recorded as KBr disk using Perkin-Elmer FT-IR spectrometer in the Faculty of Science/ School of Chemistry at University of Sulaimani.

2.2.4: 13C-NMR Spectrometry The

13

C-NMR spectra were recorded on Bruker FT-NMR spectrophotometer-500 MHz in

SAIF (Sophisticated Analytical Instrument Facility) research center in India/ Chennai, using deuterated Dimethyl sulfoxide (DMSO-d6) and deuterated Chloroform (CDCl3) as a solvent.

2.2.5: Mass Spectrometry Mass spectra were done at: -

Central research laboratory of Tehran/ Iran using Agilent Technology (HP), GC/MS model 5973 network mass selective detector.

-

Genetic and research center at University of Koya using Waters 2998 HPLC/Mass photodiode array detector.

29

2.3: Procedures: 2.3.1: Synthesis of 7-hydroxy-4-methyl Coumarin [S1] (77):

A solution of (0.01mole, 1.1 gm) resorcinol and (0.01mole, 1.3 ml) Ethyl acetoacetate was added drop wise over a period of 30 min. with stirring to (10ml) of conc. sulfuric acid in the ice bath so that the temperature of the mixture did not rise above 10 ˚C. The reaction mixture was kept at room temperature for 3 hours, and then poured with vigorous stirring into mixture of ice and water. The precipitate was filtered off and washed with water. After drying recrystallized from ethanol and creamy colored needles were obtained. The physical properties of compound [S1] are listed in table (2-2).

Table (2-2) physical properties of compound [S1]

Comp.

MP

Yield %

No.

Recrystallization

Color

solvent

TLC Solvent system

Molecular Rf

formula

value

S1

181-183

78

Ethanol

creamy

˚C

Ethylacetate:chloroform

0.68

1:9

One spot

30

C10H8O3

2.3.2: Synthesis of 8-formyl-7-hydroxy-4-methyl Coumarin [S2] (78-80):

A mixture of 7-hydroxy-4-methyl-coumarin (0.025 mole, 5.1 g) and hexamethylene tetramine (0.07 mole, 9.8 g) in glacialacetic acid (40 mL) was heated at 85-90 ˚C on water bath for 7 hr. The hexamine adduct so formed was hydrolyzed with 20% HCl (75 mL) and the mixture was heated for another 30 min. After cooling, the reaction mixture was extracted with diethyl ether (50 mL) twice. The ether layer was evaporated by using rotary evaporator and the pale yellow colored crystals obtained which were recrystallized from ethanol. The physical properties of compound [S2] are listed in table (2-3).

Table (2-3) physical properties of compound [S2]

Comp.

MP

Yield %

No.

S2

177-179

22

Recrystalliza-

Molecular

tion solvent

formula

Ethanol

C11H8O4

˚C

TLC Solvent

Rf

reagent

system

value

test

Ethylacetate:

0.727

+ve

chloroform

One

1:9

31

Tollens

spot

2.3.3: Synthesis of Aromatic Amine Schiff Bases [S3-S5] (81-83):

To a solution of (0.005) mole compound [S2] in (10 ml) absolute ethanol, appropriate aromatic amine [(0.01) mole of aniline and (0.005) mole of 4-amino benzoic acid and 4-nitro aniline] was added with continuous stirring and refluxed for (30 min). The orange colored precipitate was formed which was filtered, dried and recrystallized from ethanol. The physical properties of compound [S3-S5] are listed in table (2-4).

Table (2-4) physical properties of compound [S3-S5] Comp.

MP

No S3

168-171

Yield

Recrystalliza-

Molecular

%

tion solvent

formula

91

Ethanol

C17H13NO3

˚C

Solvent

Rf

system

value

Ethylacetate:

0.645

chloroform

one

1:9

S4

241-245

89

Ethanol

C18H13NO5

˚C

262-266

90

Ethanol

C17H12N2O5

˚C

0.82

chloroform

one

32

Bright orange

spot

Ethylacetate:

0.672

chloroform

one

1:9

Orange

spot

Ethylacetate:

1:1

S5

Color

TLC

spot

Intense orange

Rgroup

2.3.4: Synthesis of Amino Acid Schiff Bases [S6-S8] (84-86):

To a homogenous solution of (0.01 mole) amino acid (glycine, alanine and phenyl alanine) and (0.01 mole) sodium hydroxide in (20 ml) absolute ethanol; (0.01 mole) of compound [S2] which was also dissolved in absolute ethanol, was added drop by drop with continuous stirring. After 2-3 minute of the addition, 20% of the mixture was evaporated and (1 ml) of acetic acid was added. The mixture was left at room temperature for 2-3 hr. the obtained precipitate was collected by filtration, washed with cold ethanol, dried and recrystallized from suitable solvent. The physical properties of compound [S6-S8] are listed in table (2-5).

Table (2-5) physical properties of compound [S6-S8] Comp. No.

S6

MP

200-209

Yield %

72

˚C (Dec.) S7

148-151

231-237 ˚C (Dec.)

Molecular

solvent

formula

Ethanol: Water

C13H11NO5

7:3 67

˚C S8

Recrystallization

Ethanol: Water

Color

Faint yellow

C14H13NO5

yellow

C20H17NO5

Intense

7:3 53

Ethanol: Water 7:3

33

yellow

R group

2.3.5: Synthesis of Chalcones [S9-S11] (56, 87-89):

A mixture of compound [S2] (0.01 mole) and appropriate aromatic ketones [0.02 mole acetophenone, 2-hydroxy acetophenone and 0.01 mole dehydoacetic acid] was dissolved in (25 ml) absolute ethanol in a 100 ml round-bottom flask equipped with a magnetic stirrer. Then sodium hydroxide solution (10 ml, 10 %) was added drop wise to the reaction mixture on vigorous stirring for 30 minutes when the solution became turbid. The reaction temperature was maintained between 20-25 ˚C using a cold water bath on the magnetic stirrer. After vigorous stirring for 4-5 hours, the reaction mixture was left to stand for overnight at refrigerator. The precipitate obtained was filtered, washed with ice cold water, and recrystallized from suitable solvent. The physical properties of compound [S9-S11] are listed in table (2-6).

Table (2-6) physical properties of compound [S9-S11] Comp.

MP

Yield %

No. S9

170-173

Recrystallization

Molecular

solvent

formula

Color

70

Ethyl acetate

C19H14O4

Chartreuse

64

Ethyl acetate

C19H14O5

Chartreuse

36

Ethyl acetate

C19H14O7

Chartreuse

˚C S10

187-189 ˚C

S11

207-210 ˚C

34

R group

2.3.6: Synthesis of 8-(hydrazonomethyl)-7-hydroxy-4-methyl Coumarin [S12] (82) :

(0.005 mole) of compound [S2] dissolved in minimum amount of ethanol to which (0.01 mole) of 99% hydrazine hydrate was added with continuous stirring and refluxed for (30 min). A creamy colored precipitate formed which was collected by filtration and recrystallized from ethanol. The physical properties of compound [S12] are listed in table (2-7).

Table (2-7) physical properties of compound [S12]

Comp.

MP

Yield %

No. S12

201-207

91

Recrystalliza-

Molecular

tion solvent

formula

Solvent system

Rf value

Ethanol

C11H10N2O3

Ethylacetate:chloroform

0.75

1:1

one

˚C (Dec.)

TLC

spot

35

2.3.7: Synthesis of 8-((2-(2,4-dinitrophenyl)hydrazono)methyl)-7-hydroxy-4methyl Coumarin [S13] (90):

(0.005 mole) of compound [S2] and (0.005 mole) 2,4-dinitro phenyl hydrazine was refluxed in round bottom flask for 1 hr. in the presence of (20 ml) absolute ethanol as a solvent, the intense orange colored precipitate collected by filtration and recrystallized from suitable solvent. The physical properties of compound [S13] are listed in table (2-8).

Table (2-8) physical properties of compound [S13]

Comp.

MP

Yield %

No. S13

234-241

88

Recrystalliza-

Molecular

tion solvent

formula

Solvent system

Rf value

Ethanol

C17H12N4O7

Ethylacetate:chloroform

0.806

1:9

one

˚C (Dec.)

TLC

spot

36

2.3.8: Synthesis of 2-((7-hydroxy-4-methyl-2-oxo-2H-chromen-8 yl)methylene) hydrazinecarbothioamide [S14] (58, 91):

(0.005 mole) of compound [S2] and (0.005 mole) Thiosemicarbazide dissolved in (20 ml) absolute ethanol and mixed in round bottom flask with continuous stirring and refluxed for (30 min). A faint yellow colored precipitate was formed which was collected by filtration and recrystallized from ethanol. The physical properties of compound [S14] are listed in table (2-9).

Table (2-9) physical properties of compound [S14]

Comp.

MP

Yield %

No. S14

218-227

93

Recrystalliza-

Molecular

tion solvent

formula

Solvent system

Rf value

Ethanol

C12H11N3O3S

Ethylacetate:chloroform

0.503

1:9

one

˚C (Dec.)

TLC

spot

37

2.3.9: Synthesis of 2-(2-((7-hydroxy-4-methyl-2-oxo-2H-chromen-8-yl) methylene)hydrazinyl)thiazol-4(5H)-one [S15] (59, 92):

A mixture of compound [S14] (0.01 mole) and chloroacetic acid (0.01 mole) in glacial acetic acid (30 mL) containing anhydrous sodium acetate (0.04 moles) was refluxed for 17 hr. (monitored by TLC). The reaction mixture was cooled; the resulting pale yellow colored precipitate was filtered off and recrystallized from ethanol. The physical properties of compound [S15] are listed in table (2-10).

Table (2-10) physical properties of compound [S15]

Comp.

MP

Yield %

No. S15

221-234

83

Recrystalliza-

Molecular

tion solvent

formula

Solvent system

Rf value

Ethanol

C14H11N3O4S

Ethylacetate:chloroform

0.708

1:9

one

˚C (Dec.)

TLC

spot

38

Chapter Three Results and Discussion Coumarin and its derivatives are very important organic compounds possessing a wide spectrum of biological activity. They are the structural unit of several natural products. Their applications range from pharmaceuticals, optical brighteners, food additives, perfumes, cosmetics to laser dyes. Many of these compounds have proven to be active as antibacterial, antifungal, anti-inflammatory, anticoagulant, anti-HIV and antitumor agents. These properties have made coumarin interesting target for pharmaceutical chemists. 7-hydroxy-4-methyl coumarin is chosen as a starting material for the synthesis of different derivatives. The prepared derivatives include Schiff bases, chalcones, and hydrazones. All the synthesized compounds were identified by their physical properties and some spectral analysis such as FT-IR,

13

C-NMR, and MS spectroscopy and evaluated for their anti-microbial

properties. The reaction sequence of synthesized coumarin derivatives are outlined in scheme (3-1, 3-2 and 3-3) respectively.

Scheme (3-1) Synthesis of coumarin derivatives [S1 and S2] 39

Scheme (3-2) Synthesis of coumarin derivatives [S3-S11]

40

Scheme (3-3) Synthesis of coumarin derivatives [S12-S15]

41

3.1: Synthesis of 7-hydroxy-4-methyl Coumarin [S1]: Synthesis of compound [S1], scheme (3-4), was achieved by Pechmann-Duisberg reaction of ethyl acetoacetate with equimolar amount of substituted phenol; resorcinol.

Scheme (3-4) synthesis of compound [S1]

The reaction mechanism of resorcinol with ethyl acetoacetate which proceeds through electrophilic aromatic substitution using concentrated sulfuric acid to remove water and ethanol is outlined in scheme (3-5) below (10).

Scheme (3-5) mechanism of synthesis of compound [S1] The physical properties of compound [S1] are listed in table (2-2).

42

The FT-IR spectrum of compound [S1], figure (3-5), shows the O-H stretching frequency at (3155) cm-1, C=C stretching of alkene at (1678) cm-1, C=C stretching of aromatic ring at (1599) cm-1. Other significant bands were observed at (1238, 1076) that may be assigned to the C-O-C stretching frequencies. The characteristic bands of compound [S1] are listed in table (3-1).

Table (3-1) FT-IR spectral data of compound [S1] Characteristic bands of FT-IR (cm-1, KBr)

Comp. No.

[S1]

ʋ O-H

3155

ʋ C=C

ʋ C=C

alkene

aromatic

1678

1599

Others

-C-O-C (1238, 1076) - O-H bending or C-H bending of CH3 at 1450

3.2: Synthesis of 8-formyl-7-hydroxy-4-methyl Coumarin [S2]: Synthesis of compound [S2], scheme (3-6), was achieved by the Duff reaction or hexamine aromatic formylation, which is used for the synthesis of aromatic aldehyde with hexamine as the formyl carbon source, in the presence of acid (glacialacetic acid) at 85-90 ˚C, the reaction was monitored by TLC using chloroform and ethyl acetate (9:1) solvent system.

Scheme (3-6) synthesis of compound [S2]

43

The electrophilic species in this electrophilic aromatic substitution reaction is iminium ion (CH2+NR2). The initial reaction product is an iminium which is hydrolyzed to the aldehyde as shown in scheme (3-7) below (78, 79).

Scheme (3-7) mechanism of synthesis of compound [S2] 44

The reaction mechanism demonstrates how hexamine donates a methine group to an aromatic substrate via a series of equilibria reactions, with iminium ion intermediates. Initially, addition to the aromatic ring results in an intermediate at the oxidation state of a benzylamine. An intramolecular redox reaction ensues, raising the benzylic carbon to the oxidation state of an aldehyde. The oxygen atom is kindly provided by water on acid hydrolysis in the final step (93). The reaction requires strongly electron donating substituents on the aromatic ring such as in a phenol. Formylation occurs ortho to the electron donating substituent preferentially, unless the ortho positions are blocked, in which the formylation occurs in the para position (93). In case of 7-hydroxy-4-methyl coumarin formylation occur at C8 rather than C6 due to the higher resonance stability of C8 than C6 as outlined in scheme (3-8) below (94-96).

Scheme (3-8) resonance stability of 7-hydroxy-4-methyl coumarin The physical properties of compound [S2] are listed in table (2-3). 45

The FT-IR spectrum of compound [S2], figure (3-6), shows the O-H stretching frequency at (3437) cm-1, C=O stretching of ester (lactone) at (1745) cm-1, and appearance of a new band at (1644) cm-1 which is C=O stretching frequency of aldehyde with reduced frequency due to the conjugation or inter-molecular hydrogen bonding, C=C stretching of aromatic ring at (1594 and 1479) cm-1. The characteristic FT-IR bands of compound [S2] are listed in table (3-2).

Table (3-2) FT-IR spectral data of compound [S2] Characteristic bands of FT-IR (cm-1, KBr)

Comp. No.

ʋ O-H

ʋ C-H

ʋ C=O

Stretching [S2]

3437

3080 (aromatic)

2930, 2990 (aliphatic)

ʋ C=C

Others

Aromatic 1745 (lactone)

1644 (aldehyde)

1594

- OH bending at 1430

1479

-C-H bending of CH3 at 1390 -C-O-C (1228, 1072)

The 13C-NMR (ppm) spectrum of compound [S2], which is recorded in CDCl3, figure (3-20) shows the following bands of carbon: 193.399 (C=O aldehyde), 165.266 (C=O ester or lactone), 159.224, 156.14, 152.708, 132.928, 114.3, 112.055, 111.975, and 108.663 (C 18.963 (CH3). The 13C-NMR peaks of compound [S2] are listed in table (3-3).

46

aromatic and alkene),

The mass spectrum of compound [S2], figure (3-29), shows an intense molecular ion peak at m/z 204 corresponding to the molecular formula C11H8O4. The other significant bands at m/z 176, 175, 147, 143, 130 and other fragments are listed in table (3-3).

The above data are also in agreement with those published in the following research papers (79, 80, 93) .

47

Table (3-3) 13C-NMR and MS spectral data of compound [S2] C-NMR, ppm (δ)

13

Comp.

Mass spectroscopy data

No.

C=O

C aromatic and alkene

C aliphatic

[S2]

193.399

159.224, 156.14, 152.708, 132.928, 114.3, 112.055, 111.975, 108.663

18.963

204 [M] +, 176, 175, 147, 143,

(CH3)

130.

(aldehyde) 165.266

m/z

(lactone)

3.3: Synthesis of Aromatic Amine Schiff Bases [S3-S5]: The Schiff bases of aromatic amine [S3-S5], scheme (3-9), have been synthesized in good yield by condensation of compound [S2] with appropriate aromatic amines in absolute ethanol as a solvent without addition of any reagents like acid or base. The completion of the reaction and purity of the synthesized compounds were monitored by TLC.

Scheme (3-9) synthesis of compounds [S3-S5] The mechanism of reaction, as outlined in scheme (3-10), proceeds via nucleophilic attack of the lone pair of electron of amine nitrogen on the carbonyl carbon of the aldehyde with the loss of a water molecule by the application of heat (97). 48

Scheme (3-10) mechanism of synthesis of aromatic amine Schiff bases [S3-S5]

The physical properties of compound [S3-S5] are listed in table (2-4). The FT-IR spectrum of compound [S3], figure (3-7), shows a broad band of O-H stretching frequency at (3450) cm-1, C-H stretching of aromatic ring or alkene at (3080) cm-1, C-H stretching aliphatic for CH3 at (2923 and 2860) cm-1, C=O stretching of ester (lactone) at (1736) cm-1. The IR spectra also show the disappearance of C=O stretching of aldehyde at (1644) cm -1 and appearance of a new band at (1618) cm-1 which is according to the Hooke’s law (discussed below) it belongs to the C=N stretching of Schiff base. Other bands at (1577 and 1488) cm -1 belong to C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S3] are listed in table (3-4).

49

Hooke’s law which can be used to estimate the wave number of light that will be absorbed by different types of chemical bonds states that:

Ʋ bar = 1/2π * (K/μ)1/2 Where: K= force constant in (dynes/ cm), K for double bond = 10 * 105 dynes/cm μ = M1M2/ (M1+M2) (M1 and M2 are molar masses of atoms involved in the bond) According to the Hooke’s law C=N stretching frequency is nearly equal to 1627 cm-1. The 13C-NMR (ppm) spectrum of compound [S3], which is recorded in CDCl3, figure (3-21), shows the following bands of carbon: 166.877 (C=O lactone), 160.29 (C=N), 156.451, 154.55, 153.292, 146.413, 129.581, 129.317, 127.726, 121.292, 115.066, 110.985, 110.934, and 106.859 (C aromatic and alkene), 18.973 (CH3). 13C-NMR peaks of compound [S3] are listed in table (3-5).

The mass spectrum of compound [S3], figure (3-30), shows an intense molecular ion peak (M+1) at 280 corresponding to the molecular formula C17H13NO3 +1 or (M+H), and other band at m/z 202, results are listed in table (3-5).

50

The FT-IR spectrum of compound [S4], figure (3-8), shows a broad band of O-H stretching frequency at (3420) cm-1 which either belongs to the phenol group of coumarin or carboxylic acid group of the aromatic amine, C-H stretching of aromatic ring or alkene at (3070 and 3075) cm-1, C-H stretching for CH3 (aliphatic) at (2982 and 2926) cm-1, the dimer sign of carboxylic acid has appeared at nearly 2500 cm-1, C=O stretching of ester (lactone) at (1723) cm-1, C=O stretching of carboxylic acid at (1700) cm-1, C=C stretching of alkene at (1633) cm-1, C=N stretching frequency of Schiff base at (1616) cm-1. Other bands such as (1578 and 1489) are C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S4] are listed in table (3-4).

The mass spectrum of compound [S4], figure (3-31), shows an intense molecular ion peak at m/z 323 corresponding to the molecular formula C18H13NO5 . Significant bands of m/z 295, 278, 202, 175 and other fragments are listed in table (3-5).

51

The FT-IR spectrum of compound [S5], figure (3-9), shows a broad band of O-H stretching frequency at (3460) cm-1, C-H stretching of aromatic ring or alkene at (3120 and 3090) cm-1, CH stretching for CH3 (aliphatic) at (2920) cm-1, C=O stretching of ester (lactone) at (1744) cm-1, C=C stretching of alkene at (1630) cm-1, C=N stretching frequency of Schiff base at (1615) cm-1. Other bands such as (1570 and 1339) are NO2 stretching of the p-nitro group attached to the aromatic ring. The characteristic FT-IR bands of compound [S5] are listed in table (3-4). The 13C-NMR (ppm) spectrum of compound [S5], which is recorded in CDCl3, figure (3-22) shows the following bands of carbon: 165.368 (C=O lactone), 165.305 (C=N), 159.857, 154.662, 153.177, 152.99, 146.52, 132.913, 126.346, 125.277, 122.217, 114.536, 114.321, 113.355, and 112.049 (C

aromatic and alkene),

18.977 (CH3). The

13

table (3-5). 52

C-NMR peaks of compound [S5] are listed in

The mass spectrum of compound [S5], figure (3-32), shows the molecular ion peak at m/z 324 corresponding to the molecular formula C17H12N2O5 and other significant band at m/z 292; results are listed in table (3-5).

53

Table (3-4) FT-IR spectral data of compound [S3-S5] Characteristic bands of FT-IR (cm-1, KBr)

Comp. No.

[S3]

ʋ O-H

3450

ʋ C-H Stretching

3080(aromatic)

ʋ C=O

1736 (lactone)

ʋ

ʋ C=C

C=N

Aromatic

1618

1577 1488

2923& 2860

Others

- O-H bending or C-H bending of CH3 at 1388

(aliphatic) -C-O-C (1236, 1074) [S4]

3420

3070&3075

1723 (lactone)

1616

1578 1489

(aromatic)

O-H bending of carboxylic acid at 1427

1700 2982&2926

(carboxylic

O-H bending of phenol or C-H bending of CH3 at 1394

acid)

(aliphatic)

C=C alkene at 1633

-C-O-C at (1289,1077) [S5]

3460

3120&3090

1744 (lactone)

(aromatic) 2920 (aliphatic)

1615

1516

1570&1339 (NO2 stretching) O-H bending at 1390 -C-O-C at (1220,1074)

54

Table (3-5) 13C-NMR and MS spectral data of compound [S3-S5] C-NMR, ppm (δ)

13

Comp.

Mass spectroscopy

No.

C=O

C=N

C aromatic and alkene

C aliphatic

[S3]

166.877

160.29

156.451, 154.55, 153.292, 146.413, 129.581, 129.317, 127.726, 121.292, 115.066, 110.985, 110.934, 106.859.

18.973 (CH3)

280 [M+1]+ , 202.

323 [M]+, 295, 278, 202, 175.

(lactone)

[S4]

-

-

-

-

[S5]

165.368

165.305

159.857, 154.662, 153.177, 152.99, 146.52, 132.913, 126.346, 125.277, 122.217, 114.536, 114.321, 113.355, 112.049.

18.977

(lactone)

(CH3)

m/z

324 [M]+.

3.4: Synthesis of Amino Acid Schiff Bases [S6-S8]: The Schiff bases of amino acid [S6-S8], scheme (3-11), have been synthesized by reaction of compound [S2] with appropriate amino acids in absolute ethanol as a solvent by using sodium hydroxide, (NaOH) and acid, (glacial acetic acid).

55

Scheme (3-11) synthesis of compounds [S6-S8] Sodium hydroxide is used to make the NH2 group of the amino acid free since it is in resonance with the carboxylic acid group continuously. NaOH is also used to convert the carboxylic acid group of the amino acid to its sodium salt.

The mechanism of reaction as outlined in scheme (3-12), proceeds via nucleophilic attack of the lone pair of electron of amine nitrogen on the carbonyl carbon of the aldehyde with the loss of a water molecule by the use of acid; glacial acetic acid. The acid is also used to convert back the sodium salt of amino acid to carboxylic acid (86).

56

Scheme (3-12) mechanism of synthesis of amino acid Schiff bases [S6-S8] The physical properties of compound [S6-S8] are listed in table (2-5). The FT-IR spectrum of compound [S6], figure (3-10), shows a broad band of O-H stretching frequency at (3421) cm-1 which either belongs to the phenol group of coumarin or carboxylic acid group of the amino acid (glycine), C-H stretching of aromatic ring or alkene at (3070 and 3080) cm-1, C-H stretching for CH3 (aliphatic) at (2910) cm-1, C=O stretching of ester (lactone) 57

at (1724) cm-1 with reduced frequency due to the conjugation or hydrogen bonding, C=O stretching of carboxylic acid at (1650) cm-1 with reduced frequency, which might be due to intermolecular hydrogen bonding, C=N stretching frequency of Schiff base at (1582) cm-1 also with reduced frequency. Other bands such as (1531 and 1502) cm-1 are C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S6] are listed in table (3-6). The 13C-NMR (ppm) spectrum of compound [S6], which is recorded in DMSO-d6, figure (323) shows the following bands of carbon: 178.862 (C=O carboxylic acid), 172.416 (C=O lactone), 162.799 (C=N), 159.531, 157.179, 154.923, 132.077, 120.607, 106.335, 106.211, and 102.765 (C

aromatic and alkene),

53.51 (CH2), 18.147and 18.088 (CH3). The

13

C-NMR peaks of

compound [S6] are listed in table (3-7).

The mass spectrum of compound [S6], figure (3-33), shows the molecular ion peak (M+1) at 262 corresponding to the molecular formula C13H11NO5 +1 or (M+H), and other band at m/z 216 for (M- COOH). Results are listed in table (3-7).

58

The FT-IR spectrum of compound [S7], figure (3-11), shows a broad band of O-H stretching frequency at (3430) cm-1 which either belongs to the phenol group of coumarin or carboxylic acid group of the amino acid (alanine), C-H stretching of aromatic ring or alkene at (3002) cm-1, C-H stretching for CH3 (aliphatic) at (2910) cm-1, C=O stretching of (lactone) at (1712) cm-1 with reduced frequency due to the conjugation or hydrogen bonding, C=O stretching of carboxylic acid at (1642) cm-1 with reduced frequency, which might be due to intermolecular hydrogen bonding, C=N stretching frequency of Schiff base at (1581) cm-1 also with reduced frequency. Other bands such as (1520 and 1508) cm-1 are C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S7] are listed in table (3-6). The 13C-NMR (ppm) spectrum of compound [S7], which is recorded in DMSO-d6, figure (324) shows the following bands of carbon: 178.791 (C=O carboxylic acid), 175.869 (C=O lactone), 162.842 (C=N), 157.85, 157.19, 154.967, 132.074, 120.646, 106.392, 106.303, and 102.806 (C

aromatic and alkene),

50.506 (CH), 18.224 (CH3 attached to aromatic ring), 16.154 (CH3).

The 13C-NMR peaks of compound [S7] are listed in table (3-7).

The mass spectrum of compound [S7], figure (3-34), shows the molecular ion peak (M+1) at 276 corresponding to the molecular formula C14H13NO5 +1 or (M+H), and other band at m/z 230 for (M- COOH). Results are listed in table (3-7).

59

The FT-IR spectrum of compound [S8], figure (3-12), shows a broad band of O-H stretching frequency at (3434) cm-1 which either belongs to the phenol group of coumarin or carboxylic acid group of the amino acid (phenyl alanine), C-H stretching of aromatic ring or alkene at (3080 and 3085) cm-1, C-H stretching for CH3 (aliphatic) at (2930) cm-1, C=O stretching of ester (lactone) at (1716) cm-1 with reduced frequency, due to the conjugation or hydrogen bonding, C=O stretching of carboxylic acid at (1630) cm-1 with reduced frequency which might be due to intermolecular hydrogen bonding

(84)

, C=N stretching frequency of Schiff base at (1583) cm-1

also with reduced frequency. Other band such as (1503) cm-1 is C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S8] are listed in table (3-6).

60

Table (3-6) FT-IR spectral data of compound [S6-S8] Characteristic bands of FT-IR (cm-1, KBr)

Comp. No.

[S6]

ʋ O-H

3421

ʋ C-H Stretching

ʋ C=O

3080&3070

1724 (lactone)

(aromatic)

1650 (carboxylic acid)

2910(aliphatic)

ʋ

ʋ C=C

C=N

Aromatic

1582

1531 1502

Others

- O-H bending or C-H bending of CH3 at 1413 -C-O-C (1224, 1064)

[S7]

3430

3002 (aromatic)

1712 (lactone)

1581

1520 1508

2910 (aliphatic)

1642 -C-O-C at

(carboxylic

(1230,1050)

acid) [S8]

3434

3080&3085

1716 (lactone)

(carboxylic

O-H bending or C-H bending of CH3 at 1412

acid)

-C-O-C at

(aromatic) 2930 (aliphatic)

O-H bending or C-H bending of CH3 at 1412

1630

1583

1503

(1220,1073)

61

Table (3-7) 13C-NMR and MS spectral data of compound [S6-S8] C-NMR, ppm (δ)

13

Comp.

Mass spectroscopy

No.

C=O

C=N

C aromatic and alkene

C aliphatic

[S6]

178.862

162.799

159.531, 157.179, 154.923, 132.077, 120.607, 106.335, 106.211, 102.765.

53.51

(carboxy lic acid)

[S7]

18.147& 18.088

(lactone)

(CH3) 162.842

(carboxy lic acid)

[S8]

(CH2) C-N 262 [M+1]+, 216.

172.416

178.791

157.85, 157.19, 154.967, 132.074, 120.646, 106.392, 106.303, 102.806.

50.506 (CH) C-N

6.154

(lactone)

(CH3) -

-

276 [M+1]+, 230.

18.224&1

175.869

-

m/z

-

-

3.5: Synthesis of Chalcones [S9-S11]: The chalcones or α, β unsaturated ketones [S9-S11], scheme (3-13), are synthesized by reaction of compound [S2] with appropriate aromatic ketones by using ethanol as a solvent and using sodium hydroxide (NaOH) as a reagent.

62

Scheme (3-13) synthesis of compounds [S9-S11]

The mechanism of the reaction proceeds via aldol condensation reaction in which, an enol or an enolate ion reacts with a carbonyl compound (8-formyl-7-hydroxy-4-methyl coumarin) to form a β-hydroxyketone, followed by a dehydration to give a conjugated enone (α, β unsaturated ketones). The mechanism of the reaction consists of four main steps as outlined in scheme (3-14) (98): -

Step one is an acid-base reaction, hydroxide functions as a base and removes an acidic αhydrogen giving a carbanion intermediate or reactive enolate.

-

Step two is alkoxide formation; the nucleophilic enolate attacks the carbonyl carbon of 8formyl-7-hydroxy-4-methyl coumarin in a nucleophilic addition process giving an intermediate alkoxide.

-

Step three is protonation of alkoxide, the alkoxide deprotonates a water molecule producing β-hydroxyketone.

-

Step four is dehydration an elimination reaction in which a water molecule is removed by the action of base leading to the final product α, β unsaturated ketones.

63

Scheme (3-14) mechanism of synthesis of compounds [S9-S11]

The physical properties of compound [S9-S11] are listed in table (2-6). The FT-IR spectrum of compound [S9], figure (3-13), shows a broad band of O-H stretching frequency at (3394) cm-1 , C-H stretching frequency of aromatic ring or alkene at (3080) cm-1, CH stretching for CH3 (aliphatic) at (2922 and 2870) cm-1, C=O stretching of ester (lactone) at 64

(1719) cm-1 with reduced frequency, due to the conjugation or hydrogen bonding, C=O stretching of ketone at (1684) cm-1 with reduced frequency due to the conjugation (α,β unsaturated), C=C stretching frequency of conjugated alkene at (1640) cm-1 also with reduced frequency. Other band such as (1584) cm-1 is C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S9] are listed in table (3-8). The FT-IR spectrum of compound [S10], figure (3-14), shows a broad band of O-H stretching frequency at (3420) cm-1 , C-H stretching frequency of aromatic ring or alkene at (3095 and 3005) cm-1, C-H stretching for CH3 (aliphatic) at (2950 and 2890) cm-1, C=O stretching of ester (lactone) at (1719) cm-1 with reduced frequency due to the conjugation or hydrogen bonding, C=O stretching of ketone at (1685) cm-1 with reduced frequency due to the conjugation (α,β unsaturated), C=C stretching frequency of conjugated alkene at (1640) cm-1 also with reduced frequency. Other band such as (1585) cm-1 is C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S10] are listed in table (3-8). The 13C-NMR (ppm) spectrum of compound [S10], which is recorded in DMSO-d6, figure (325) shows the following bands of carbon: 190.705 (C=O α,β unsaturated ketone) (C=O lactone), 164.301, 157.049, 155.991, 131.438(C

aromatic),

121.626 (C

(88)

, 178.226

aromatic and alkene),

111.525, 106.392, 105.306 (C aromatic), 18.255 (CH3). The 13C-NMR peaks of compound [S10] are listed in table (3-9).

65

The mass spectrum of compound [S10], figure (3-35), shows the molecular ion peak at m/z 322 corresponding to the molecular formula C19H14O5. Other significant bands at m/z 201, 175 and other fragments are listed in table (3-9).

The FT-IR spectrum of compound [S11], figure (3-15), shows a broad band of O-H stretching frequency at (3393) cm-1 , C-H stretching frequency of aromatic ring or alkene at (3040) cm-1, C-H stretching for CH3 (aliphatic) at (2960 and 2910) cm-1, C=O stretching of ester (lactone) at (1716) cm-1 with reduced frequency due to the conjugation or hydrogen bonding. Both (1685) cm-1 are C=O stretching of ketones with reduced frequency, due to the conjugation (α,β unsaturated) and intermolecular hydrogen bonding, C=C stretching frequency of conjugated alkene at (1611 and 1640) cm-1. Other band such as (1584) cm-1 is C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S11] are listed in table (3-8).

66

The 13C-NMR (ppm) spectrum of compound [S11], which is recorded in DMSO-d6, figure (326) shows the following bands of carbon: 190.905 (C=O α,β unsaturated ketone) (C=O lactone), 165.036, 164.446, 157.215, 156.076 (C 111.638, 106.557, 105.422 (C

aromatic and alkene),

aromatic),

131.598 (C=C

(88)

alkene),

, 178.312 121.634,

37.871, 37.704 (CH3), 18.299 (CH3). The

13

C-

NMR peaks of compound [S11] are listed in table (3-9).

The mass spectrum of compound [S11], figure (3-36), shows the molecular ion peak at m/z 354 corresponding to the molecular formula C19H14O7. Other significant bands at m/z 201, 175 and other fragments are listed in table (3-9).

67

68

Table (3-8) FT-IR spectral data of compound [S9-S11] Characteristic bands of FT-IR (cm-1, KBr)

Comp. No.

ʋ O-H

ʋ C-H Stretching

ʋ C=O

ʋ C=C

Others

[S9]

3394

3080 (aromatic or

1719 (lactone)

1640 (alkene)

1481 (C-H bending of CH2)

1684 (ketone)

1584 (aromatic)

alkene) 2922&2870

1434 (C-H bending of CH3)

(aliphatic) 1416 (O-H bending) 1220&1072 (C-O-C) [S10]

3420

3095&3005

1719 (lactone)

1640 (alkene)

1685 (ketone)

1585 (aromatic)

(aromatic or alkene)

1481 (C-H bending of CH2) 1435 (C-H bending of CH3)

2950&2890 1416 (O-H bending)

(aliphatic)

1220&1072 (C-O-C) [S11]

3393

3040 (aromatic or

1716 (lactone)

alkene) 2960&2910

1611and 1640 (alkene)

1685 (ketone) 1584 (aromatic)

(aliphatic)

1483 (C-H bending of CH2) 1436 (C-H bending of CH3) 1415 (O-H bending) 1222&1072 (C-O-C)

69

Table (3-9) 13C-NMR and MS spectral data of compound [S9-S11] C-NMR, ppm (δ)

13

Comp.

Mass spectroscopy

No.

C=O

C=C

C aromatic and alkene

C aliphatic

m/z

[S9]

-

-

-

-

-

[S10]

190.705

121.626

164.301, 157.049, 155.991, 131.438, 121.626, 111.525, 106.392, 105.306.

18.255

165.036, 164.446, 157.215, 156.076, 121.634, 111.638, 106.557, 105.422.

37.871&

(ketone) 178.226

322 [M]+, 201, 175.

(CH3)

(lactone) [S11]

190.905 (ketone) 178.312

131.598

(lactone)

354 [M]+, 201, 175.

37.704 (CH3) 18.299 (CH3)

3.6: Synthesis of Hydrazone Derivatives [S12-S14]: The hydrazone derivatives of coumarin [S12-S14], scheme (3-15), have been synthesized in a good yield by condensation of compound [S2] with hydrazine hydrate 99% [S12], 2,4-di-nitro phenyl hydrazine [S13], and thiosemicarbazide [S14] in absolute ethanol as a solvent by the action of heat without addition of any reagents like acid or base.

70

Scheme (3-15) synthesis of compounds [S12-S14] The mechanism of reaction as outlined in scheme (3-16), proceeds via nucleophilic attack of the lone pair of electron of hydrazine nitrogen (which is more nucleophilic than a regular amine due to the presence of the adjacent nitrogen) on the carbonyl carbon of the aldehyde with the loss of a water molecule by the action of heat (83).

Scheme (3-16) mechanism of synthesis of hydrazone derivatives [S12-S14] 71

The physical properties of compound [S12-S14] are listed in table (2-7, 8 and 9). The FT-IR spectrum of compound [S12], figure (3-16), shows a sharp band of O-H stretching frequency at (3381) cm-1 which indicates that the O-H group is free, NH2 stretching frequency gave a double peak at (3295 and 3218) cm1, C-H stretching for CH3 at (2910 and 2865) cm-1, C=O stretching of ester (lactone) at (1695) cm-1 with reduced frequency due to the conjugation or intermolecular hydrogen bonding, C=N stretching frequency at (1605) cm-1. Other bands such as (1579) and (1491) cm-1 belong to N-H bending and C=C stretching of aromatic ring, respectively. The characteristic FT-IR bands of compound [S12] are listed in table (3-10). The FT-IR spectrum of compound [S13], figure (3-17), shows a broad band of O-H stretching frequency at (3420) cm-1, N-H stretching frequency at (3231) cm1, C-H stretching frequency of aromatic ring or alkene at (3095) cm-1, C-H stretching for CH3 at (2910) cm-1, C=O stretching of ester (lactone) at (1736) cm-1, C=C stretching frequency of alkene at (1640) cm-1, C=N stretching frequency at (1607) cm-1. Other bands such as (1590) cm-1 C=C stretching of aromatic ring, N-H bending frequency at (1550) cm-1 and NO2 stretching at (1519 and 1386) cm-1. The characteristic FT-IR bands of compound [S13] are listed in table (3-10). The mass spectrum of compound [S13], figure (3-37), shows an intense molecular ion peak at m/z 384 corresponding to the molecular formula C17H12N4O7, and other significant bands at m/z 367, 323, 202, 188, 175 and other fragments are listed in table (3-11).

72

The FT-IR spectrum of compound [S14], figure (3-18), shows a broad band of O-H stretching frequency which is mixed with NH2 and N-H stretching region at nearly (3400- 3200) cm-1, NH2 stretching frequency gave a double peak at (3390 and 3283) cm1 N-H stretching at (3185) cm-1, C-H stretching of aromatic or alkene at (3010)cm-1, C=O stretching of ester (lactone) at (1727) cm-1 with reduced frequency, C=N stretching frequency at (1599) cm-1. Other bands such as (1524 and 1492) cm-1 belong to N-H bending and C=C stretching of aromatic ring respectively. The characteristic FT-IR bands of compound [S14] are listed in table (3-10). The 13C-NMR (ppm) spectrum of compound [S14] which is recorded in DMSO-d6, figure (327) shows the following bands of carbon: 178.319 (C=O lactone), 164.141 (C=N), 160.034 (C=S), 159.728, 159.232, 154.075, 153.922, 133.729, 121.697, 113.667, 112.572, and 111.14

73

(C

aromatic and alkene),

55.551 (C-N), 18.791 (CH3). The

13

C-NMR peaks of compound [S14] are

listed in table (3-11).

The mass spectrum of compound [S14], figure (3-38), shows intense molecular ion peak at m/z 277 corresponding to the molecular formula C12H11N3O3S. Other significant bands at m/z 202, 188, 175 and other fragments are listed in table (3-11).

74

Table (3-10) FT-IR spectral data of compound [S12-S14] Characteristic bands of FT-IR (cm-1, KBr)

Comp. No.

[S12]

ʋ O-H

3381

ʋ N-H

ʋ C-H

Stretching

Stretching

3295&3218

2910&2865 (aliphatic)

(NH2)

ʋ C=O

ʋ C=N

Others

1695

1605

1579 (N-H bending) 1491 (C=C aromatic) 1385 (O-H or C-H bending of CH3) 1228& 1072 (C-O-C)

[S13]

3420

3231 (N-H)

3095 (aromatic or alkene)

1736

1607

1640 (C=C alkene) 1590 (C=C aromatic) 1550 (N-H bending)

2910 (aliphatic)

1519&1386 (NO2) stretching 1226& 1068 (C-O-C)

[S14]

3400-

3390&3283

3010

3200

(NH2)

(aromatic or alkene)

3185 (N-H)

1727

1599

1524 (N-H bending) 1492 (C=C aromatic) 1431(O-H bending) 1217& 1080 (C-O-C)

75

Table (3-11) 13C-NMR and MS spectral data of compound [S12-S14] C-NMR, ppm (δ)

13

Comp.

Mass spectroscopy

No.

C=O

C=N

C aromatic and alkene

others

m/z

[S12]

-

-

-

-

-

[S13]

-

-

-

-

384 [M]+, 367, 323, 202, 188, 175.

[S14]

178.319 (lactone)

164.141

159.728, 159.232, 154.075, 153.922, 133.729, 121.697, 113.667, 112.572, 111.14.

160.034 (C=S) 55.551 (C-N) 18.791 (CH3).

76

277 [M]+, 202, 188, 175.

3.7: Synthesis of 2-(2-((7-hydroxy-4-methyl-2-oxo-2H-chromen-8-yl) methylene)hydrazinyl)thiazol-4(5H)-one [S15]: Synthesis of compound [S15], scheme (3-17), was achieved by condensation of compound [S14] with chloroacetic acid in the presence of sodium acetate in glacial acetic acid as a solvent.

Scheme (3-17) synthesis of compound [S15]

77

The mechanism of the reaction is outlined in scheme (3-18) below (92):

Scheme (3-18) mechanism of synthesis of compound [S15] The physical properties of compound [S15] are listed in table (2-10).

78

The FT-IR spectrum of compound [S15], figure (3-19), shows a band of O-H stretching frequency at (3446) cm-1. Disappearance of NH2 band while N-H stretching frequency appeared at nearly (3300) cm1, C-H stretching of aromatic or alkene at (3000)cm-1, C-H stretching of CH3 at (2930) cm-1, C=O stretching of ester (lactone) at (1731) cm-1, the appearance of new band of C=O stretching frequency of amide at (1687) cm-1, C=N stretching frequency at (1624) cm-1, NH bending at (1561) cm-1. Other bands (1520) cm-1 demonstrates C=C stretching of aromatic ring. The characteristic FT-IR bands of compound [S15] are listed in table (3-12).

Table (3-12) FT-IR spectral data of compound [S15] Characteristic bands of FT-IR (cm-1, KBr)

Comp. No.

[S15]

ʋ O-H

3446

ʋ N-H

ʋ C-H

Stretching

Stretching

3300

3000 (aromatic or alkene) 2930 (aliphatic)

ʋ C=O

ʋ C=N

Others

1731 (lactone)

1624

1561 (N-H bending)

1687 (amide)

1520 (C=C aromatic) 1385 (O-H or C-H bending of CH3) 1239-1085 (C-O-C)

The 13C-NMR (ppm) spectrum of compound [S15], which is recorded in DMSO-d6, figure (328) shows the following bands of carbon: 174.162 (C=O amide), 166.609 (C=O lactone), 161.939 (C=N), 159.827 (C

aromatic

attached to OH group), 159.497 (C=N), 154.212, 153.432,

129.445, 116.611, 113.672, 112.448, 112.4 (C aromatic and alkene), 34.137 (C-S), 18.792 (CH3). The 13

C-NMR peaks of compound [S15] are listed in table (3-13).

79

The mass spectrum of compound [S15], figure (3-39), shows an intense molecular ion peak at m/z 317 corresponding to the molecular formula C14H11N3O4S. Other significant bands at m/z 202, 175, 160 and other fragments are listed in table (3-13).

80

Table (3-13) 13C-NMR and MS spectral data of compound [S15] C-NMR, ppm (δ)

13

Comp.

Mass spectroscopy

No.

C=O

C=N

C aromatic and alkene

others

m/z

[S15]

174.162

161.939

159.827, 154.212, 153.432, 129.445, 116.611, 113.672, 112.448, 112.4.

34.137 (C-S)

317 [M]+, 202, 175,

(amide) 166.609 (lactone)

159.497

18.792

160.

(CH3).

3.8: Anti-microbial Activity: Antimicrobial agents are chemicals that kill or inhibit the growth of micro-organisms and are used to treat microbial infections. Some are produced naturally by microbes but many are synthetic. Antimicrobials include antibiotics, antivirals, antifungals and other natural plant bioactive compounds (99).

3.8.1: Antibacterial Susceptibility Tests: Antibacterial susceptibility testing may be performed by either dilution or diffusion methods. The choice of methodology is often based on many factors, including relative ease of performance, flexibility, use of automated or semi-automated devices for both identification and susceptibility testing (100). There are many methods implied to test antimicrobial activity of chemical compounds. Standard laboratory procedures which are applied are three main methods that are still used by standard institutes for setting guidelines. Conventional methods of testing antimicrobial activities are broth dilution, agar dilution and disc diffusion methods (101).

3.8.1.1: Dilution Methods: Dilution susceptibility testing methods are used to determine the minimal concentration, usually expressed in micrograms per milliliter of antimicrobial agents required to inhibit the 81

growth of a microorganism. Procedures for determining the antimicrobial inhibitory activity are carried out by either agar or broth based methods. Antimicrobial agents are usually tested as (twofold) serial dilutions, but any other concentrations can be set in between. The lowest concentration that inhibits visible growth of an organism in vitro after overnight incubation is recorded as the MIC (101). Minimum inhibitory concentrations (MICs) are considered the gold standard for determining the susceptibility of organisms to antimicrobials and are, therefore, used to judge the performance of all other methods of susceptibility testing. Minimum inhibitory concentrations are important in diagnostic laboratories to confirm resistance of microorganisms to an antimicrobial agent. MIC is generally regarded as the most basic laboratory measurement of the activity of an antimicrobial agent against an organism, which provides a quantitative result (102,103)

.

The general approaches for broth methods include macro-broth dilution, in which the broth volume for each antimicrobial concentration is ≥ 1.0 ml contained in test tubes and micro-broth dilution, in which antimicrobial dilutions are in 0.05 to 0.1 ml volumes contained in wells of micro-titer trays. The macro-broth dilution broth method is a well standardized and reliable reference method that is useful for research purposes. However, because of the laborious nature of the procedure and the availability of more convenient dilution systems (micro dilution), this procedure is generally not useful for routine susceptibility testing in clinical laboratories (102).

3.8.1.2: Disk Diffusion Method: Disc diffusion tests were introduced mainly because they were less cumbersome technically when large numbers of organisms were tested against many antimicrobials including antibiotics. The disk diffusion method of susceptibility testing allows categorization of bacterial isolates as susceptible, resistant, or intermediate to a variety of antimicrobial agents. It is technically simple to perform and very reproducible. Moreover, it does not require any special equipment and provides category results that are easily interpreted by clinicians. The primary disadvantage of disk diffusion susceptibility testing is that, it provides a qualitative result (103,104). In the present study the macro-broth dilution testing method is performed for screening antibacterial activity of new coumarin derivatives against two Gram positive (G+ve) bacteria 82

Staphylococcus epidermidis and Staphylococcus hemolyticus, and two Gram negative (G-ve) bacteria Escherichia coli and Klebsiella pneumoniae to determine their MIC in vitro. A series of 6 to 9 broth tubes containing 1 mL Muller Hinton autoclaved and cooled then inoculated with (50 µL) bacterial inoculum of bacterial suspension at McFarland turbidity of 0.5. Then a defined concentration of coumarin derivatives are added to each broth of a serial doubling concentration except the negative control in which the solvent (DMSO) was added and a positive control with bacterial inoculum without adding any derivatives. After those broths are incubated aerobically at 37 ˚C for 24 hour, MIC is determined by visual observation as the lowest concentration that inhibits bacterial growth and appears as clear broth tube. All the synthesized compounds except compound [S1] are tested for their anti-microbial (antibacterial) activities. Results are listed in table (3-14). Table (3-14) Anti-bacterial activity of compound [S2-S15] Compound

Staphylococcus

Staphylococcus

Escherichia

Klebsiella

No.

epidermidis (G+ve)

hemolyticus (G+ve)

coli (G-ve)

pneumoniae (G-ve)

S2

100 µg/mL

100 µg/mL

150 µg/mL

200 µg/mL

S3

100 µg/mL

75 µg/mL

100 µg/mL

150 µg/mL

S4

150 µg/mL

100 µg/mL

100 µg/mL

150 µg/mL

S5

100 µg/mL

75 µg/mL

150 µg/mL

150 µg/mL

S6

75 µg/mL

100 µg/mL

100 µg/mL

150 µg/mL

S7

50 µg/mL

100 µg/mL

100 µg/mL

150 µg/mL

S8

50 µg/mL

75 µg/mL

75 µg/mL

150 µg/mL

S9

75 µg/mL

75 µg/mL

25 µg/mL

100 µg/mL

S10

50 µg/mL

50 µg/mL

75 µg/mL

150 µg/mL

S11

100 µg/mL

100 µg/mL

100 µg/mL

150 µg/mL

S12

50 µg/mL

25 µg/mL

50 µg/mL

150 µg/mL

S13

75 µg/mL

75 µg/mL

100 µg/mL

100 µg/mL

S14

100 µg/mL

100 µg/mL

100 µg/mL

150 µg/mL

S15

50 µg/mL

50 µg/mL

100 µg/mL

100 µg/mL

83

From the results illustrated in table (3-14) it is clear that all the synthesized coumarin derivatives have anti-bacterial activity against both gram positive and gram negative bacteria in vitro, the MIC ranges between 25-200 µg/mL in which they varies according to the derivatives and bacteria. The lowest MIC is 25 µg /mL for both compounds S9 against Escherichia coli and S12 against Staphylococcus hemolyticus, and the highest MIC of 200 µg /mL for compound S2 against Klebsiella pneumoniae, figures (3-1 and 3-2) shows anti-bacterial activity of some of the synthesized compounds.

Figure (3-1) Anti-bacterial activity of compound [S9] against Escherichia coli

84

Figure (3-2) Anti-bacterial activity of compound [S15] against Klebsiella pneumoniae According to the statistical analysis and the mean values, table (3-15), the synthesized coumarin derivatives have greater activity against G+ve bacteria than G-ve bacteria.

Table (3-15) Mean values of MIC

Mean MIC

Staphylococcus

Staphylococcus

Escherichia

Klebsiella

epidermidis (G+ve)

hemolyticus (G+ve)

coli (G-ve)

pneumoniae (G-ve)

80.35 µg/mL

78.57 µg/mL

94.64 µg/mL

142.85 µg/mL

Generally, the MIC of all synthesized compounds is higher for Klebsiella pneumoniae compared to other bacteria which might be due to its prominent poly saccharide capsule (which is composed of 63% capsular polysaccharide, 30% lipopolysaccharide, and 7% protein) around bacterial cell which may play a protective role for the bacteria (105). Disk diffusion method for antimicrobial activity has been tried for above coumarin derivatives. However, due to the lack of diffusion of almost all of compounds over the petridish agar media rendered this technique not useful method for testing their anti-bacterial activity, and this derived us to use serial broth dilution method to test their anti-bacterial activity, as illustrated in figure (3-3) below. 85

(a)

(b)

Figure (3-3) Disc diffusion method of anti-bacterial susceptibility test a- Disc diffusion method of synthesized coumarin derivatives showing no inhibition zone. b- Disc diffusion method of synthesized coumarin derivatives with standard 10 µg Imipenem disc, with clear zone of inhibition against Escherichia coli

While direct drops containing 50-200 µg of compounds which was placed over the inoculated petridish that spread out about 1cm over the stricken bacterial inoculum showed inhibited zone of growth. It gives a clue of having antibacterial activity over the area of direct contact but not beyond drop contact. It also comes back to mind that minimal or lack of diffusion or solubility of chemical compounds beyond border of drop contact, as shown in figure (3-4).

86

Figure (3-4) Anti-bacterial activity of some coumarin derivatives

87

Figure (3-5) FT-IR spectrum of compound [S1]

Figure (3-6) FT-IR spectrum of compound [S2]

88

Figure (3-7) FT-IR spectrum of compound [S3]

Figure (3-8) FT-IR spectrum of compound [S4]

89

Figure (3-9) FT-IR spectrum of compound [S5]

Figure (3-10) FT-IR spectrum of compound [S6]

90

Figure (3-11) FT-IR spectrum of compound [S7]

Figure (3-12) FT-IR spectrum of compound [S8]

91

Figure (3-13) FT-IR spectrum of compound [S9]

Figure (3-14) FT-IR spectrum of compound [S10]

92

Figure (3-15) FT-IR spectrum of compound [S11]

Figure (3-16) FT-IR spectrum of compound [S12]

93

Figure (3-17) FT-IR spectrum of compound [S13]

Figure (3-18) FT-IR spectrum of compound [S14]

94

Figure (3-19) FT-IR spectrum of compound [S15]

95

Figure (3-20) 13C-NMR spectrum of compound [S2]

Figure (3-21) 13C-NMR spectrum of compound [S3] 96

Figure (3-22) 13C-NMR spectrum of compound [S5]

Figure (3-23) 13C-NMR spectrum of compound [S6] 97

Figure (3-24) 13C-NMR spectrum of compound [S7]

Figure (3-25) 13C-NMR spectrum of compound [S10] 98

Figure (3-26) 13C-NMR spectrum of compound [S11]

Figure (3-27) 13C-NMR spectrum of compound [S14] 99

Figure (3-28) 13C-NMR spectrum of compound [S15]

A

b u

n d

a n

c

e

S 1 8

0 0

0 0

0

1 7

0 0

0 0

0

1 6

0 0

0 0

0

1 5

0 0

0 0

0

1 4

0 0

0 0

0

1 3

0 0

0 0

0

1 2

0 0

0 0

0

1 1

0 0

0 0

0

1 0

0 0

0 0

0

9 0

0 0

0 0

8 0

0 0

0 0

7 0

0 0

0 0

6 0

0 0

0 0

5 0

0 0

0 0

4 0

0 0

0 0

3 0

0 0

0 0

2 0

0 0

0 0

c

a

n

1

4

( 0 . 2 8 1

) :

6

4 0

1 5

1 2

1 4

8 . D

8

1 7

6

9 1

5 5

6 5

7 7 1 1

0 0

in

2 0

4 3

1 0

m

8 4

1 0 9 8

0 0

9

5

1 3 1 1

0

2

1 5 1 3

1 6

5

1 8

7

9

3 1 9

0

1 9

0

1 9

7

0 4 0 m

/

5 0

6 0

7 0

8 0

9 0

1 0

0

1 1

0

1 2

0

1 3

0

1 4

0

1 5

0

1 6

0

z - - >

Figure (3-29) Mass spectrum of compound [S2]

100

1 7

0

1 8

0

2 0

0

4

Figure (3-30) Mass spectrum of compound [S3]

101

A b u n d a n c e

S c a n 3 0 0 0 0

4 4 7

(2 .3 0 6

m in ) : 6 4 0 1 5 1 2 9 . D

4 5 6 5

2 5 0 0 0

7 7

2 0 0 0 0

1 5 0 0 0 9 1

3 2 3

1 0 0 0 0 2 9 5 1 0 3 5 0 0 0

1 2 1

1 7 2

1 4 6

1 3 3

1 5 8

1 8 7

2 0 2 2 2 02 3 2

2 5 0 2 6 62 7 8

0 4 0

6 0

8 0

1 0 0

1 2 0

1 4 0

1 6 0

1 8 0

2 0 0

2 2 0

2 4 0

m / z -->

Figure (3-31) Mass spectrum of compound [S4]

Figure (3-32) Mass spectrum of compound [S5]

102

2 6 0

2 8 0

3 0 0

3 2 0

Figure (3-33) Mass spectrum of compound [S6]

103

Figure (3-34) Mass spectrum of compound [S7]

104

Figure (3-35) Mass spectrum of compound [S10]

Abundanc e S c a n 3 3 3 (1 . 7 6 4 m in ): 6 4 0 1 5 1 3 4 . D 14000

43 63

12000 10000

91 77

8000 6000 4000 105 119

2000

147

172 202 188

0 40

60

80

232

266

289

335 354

100 120 140 160 180 200 220 240 260 280 300 320 340

m / z -->

Figure (3-36) Mass spectrum of compound [S11]

105

A

b

u

n

d

a

n

c

e

S 6

2

5

0

0

0

2

4

0

0

0

2

3

0

0

0

2

2

0

0

0

2

1

0

0

0

2

0

0

0

0

1

9

0

0

0

1

8

0

0

0

1

7

0

0

0

1

6

0

0

0

1

5

0

0

0

1

4

0

0

0

1

3

0

0

0

1

2

0

0

0

1

1

0

0

0

1

0

0

0

0 4

9

0

0

0

8

0

0

0

7

0

0

0

6

0

0

0

5

0

0

0

4

0

0

0

3

0

0

0

2

0

0

0

1

0

0

0

c

a

n

7

5

6

( 3

. 7

7

5

m

in

) :

6

4

0

1

5

1

3

0

. D

3 7

7

9

1

1

7

2

2

0

2

1

3

1

1

8

4

8

1

1

3

4

6

2

2 1

8

2

0

8

3 2

3

4

2

4

2

4

8 2

6

2

0

5

2

8

0 2

9

5

2

8

0

3

0

2

3 3

4

0

3

4

0

3

6

7

0 4 m

/

0

6

0

8

0

1

0

0

1

2

0

1

4

0

1

6

0

1

8

0

2

0

0

2

2

0

0

6

0

3

2

0

3

6

z - - >

Figure (3-37) Mass spectrum of compound [S13] A b u n d a n c e

S c a n

3 9 6

(2 .0 6 4

m in ) :

6 4 0 1 5 1 3 1 .D

5 1

3 0 0 0 0 2 8 0 0 0 2 6 0 0 0 4 1 2 4 0 0 0

6 3

2 2 0 0 0 2 0 0 0 0 7 7

1 8 0 0 0

8 9

1 7 3

1 6 0 0 0 1 4 0 0 0 1 2 0 0 0 1 0 0 0 0 1 0 3

8 0 0 0 6 0 0 0

1 1 7

4 0 0 0

2 0 2 1 3 1

1 4 7 1 6 0

2 0 0 0

1 8 9

2 1 8

2 3 92 4 9

2 6 62 7 7

0 4 0

6 0

8 0

1 0 0

1 2 0

1 4 0

1 6 0

1 8 0

2 0 0

2 2 0

m / z -->

Figure (3-38) Mass spectrum of compound [S14] 106

2 4 0

2 6 0

0

3

8

0

A b u n d a n c e

S c a n

3 9 0

(2 . 0 3 5

m

in ) :

6 4 0 1 5 1 3 2 . D

7 5 0 0 0 4 2 7 0 0 0 0 6 5 0 0 0 6 0 0 0 0 5 5 0 0 0 5 0 0 0 0 4 5 0 0 0 4 0 0 0 0 6 3

3 5 0 0 0 3 0 0 0 0 2 5 0 0 0 2 0 0 0 0

1 3 1

1 6 0

1 5 0 0 0

3 1 7

1 0 0 0 0 7 7 5 0 0 0

1 4 5 1 7 2

9 1 1 0 3 1 1 7

1 8 9

2 0 2

0 4 0 m

6 0

8 0

1 0 0

1 2 0

1 4 0

1 6 0

1 8 0

2 0 0

2 2 6 2 2 0

/ z -->

Figure (3-39) Mass spectrum of compound [S15]

107

2 9 2

2 4 82 6 0 2 4 0

2 6 0

2 8 0

3 0 0

Chapter Four Conclusions and Recommendations

4.1: Conclusions: In the present study some new coumarin derivatives have been synthesized from 7-hydroxy-4methyl coumarin by applying Duff reaction to insert the formyl group to carbon 8 of coumarin nucleus. The synthesized compounds include Schiff bases, Chalcones, hydrazones, and cyclized thiosemicarbazone derivative. The compounds [S2-S15] have been evaluated for anti-bacterial activity by determining their minimum inhibitory concentration (MIC) by serial broth dilution method, in which they demonstrated antibacterial activity against gram positive more than the gram negative bacteria with varying ranges of MIC.

4.2: Recommendations: 1- Further spectral analysis such as 1H-NMR and CHN analysis. 2- Antifungal activity of the synthesized compounds. 3- Animal and cell line studies of the synthesized compounds for determining anti-cancer, antioxidant and anti-inflammatory activity and studying their pharmacokinetic properties.

108

References 1.

Alvarez-Builla J, Vaquero J, Barluenga J. Modern Heterocyclic Chemistry: Wiley-VCH Verlag GmbH & Co. KGaA.; 2011.

2.

Hauptmann TE. The Chemistry of Heterocyclic Compounds. Wiley interscience, New York. 2008;49.

3.

Katritzky AR, Drum CA. Comprehensive Heterocyclic Chemistry. Pergamon Press, Oxford. 1984;1:47.

4.

Katritzky AR, Rees CW, Scriven FV. Comprehensive Heterocyclic Chemistry II. Pergamon Press, Oxford. 1996;3:229.

5.

Ellis GP. The Chemistry of Heterocyclic Compounds, Chromenes, Chromanones, and Chromones. 1977;31.

6.

Lake BG. Coumarin metabolism, toxicity and carcinogenicity: relevance for human risk assessment. Food and Chemical Toxicology. 1999;37:423-53.

7.

Ojala T. Biological screening of plant coumarins. PhD thesis. University of Helsinki; 2001.

8.

Lacy A, O’Kennedy R. Studies on coumarins and coumarin-related compounds to determine their therapeutic role in the treatment of cancer. Current Pharmaceutical Design. 2004;10:3797-811.

9.

Heravi M, Khaghaninejad S, Mostofi M. Pechmann reaction in the synthesis of coumarin derivatives. Advances in Heterocyclic Chemistry. 2014;112.

10.

Tyagi B, Mishra MK, Jasra RV. Synthesis of 7-substituted 4-methyl coumarins by Pechmann reaction using nano-crystalline sulfated-zirconia. Journal of Molecular Catalysis A: Chemical. 2007;276:47–56.

11.

Chakravarti D. Synthesis of coumarins and chromones. Calcutta: University College of Science and Technology, 1938;5(2).

12.

Sahoo S, Shukla S, Nandy S, Sahoo HB. Synthesis of novel coumarin derivatives and its biological evaluations. European Journal of Experimental Biology. 2012;2(4):899-908.

13.

Augustine JK, Bombrun A, Ramappa B, Boodappa C. An efficient one-pot synthesis of coumarins mediated by propylphosphonic anhydride (T3P) via the Perkin condensation. Tetrahedron Letters. 2012;53:4422–5.

109

14.

Crawford M, Shaw J. The course of the Perkin coumarin synthesis. Part I. Journal of the Chemical Society. 1953:3435-9.

15.

Borges F, Roleira F, Milhazes N, Santana L, Uriarte E. Simple coumarins and analogues in medicinal chemistry: Occurrence, synthesis and biological activity. Current Medicinal Chemistry. 2005;12(8):887-916.

16.

Shaabani A, Ghadari R, Rahmati A, Rezayan AH. Coumarin synthesis via Knoevenagel condensation reaction in 1,1,3,3-N,N,N',N'-tetramethylguanidinium trifluoroacetate ionic liquid. Journal of The Iranian Chemical Society. 2009;6(4):710-4.

17.

Valizadeh H, Vaghefi S. One-Pot Wittig and Knoevenagel reactions in ionic liquid as convenient

methods

for

the

synthesis

of

coumarin

derivatives.

Synthetic

Communications. 2009;39(9):1666-78. 18.

Irgashev RA, Karmatsky AA, Slepukhin PA, Rusinov GL, Charushin VN. A convenient approach to the design and synthesis of indolo[3,2-c]coumarins via the microwaveassisted Cadogan reaction. Tetrahedron Letters. 2013;54:5734–8.

19.

Bezwada RS. Chemistry of coumarins. Hillsborough: INDOFINE Chemical Company; 2008.

20.

HamaSalih M. Synthesis of some fused heterocyclic compounds from Hymechromone. MSc Thesis. University of Mosul; 2007.

21.

Rabek JF, Scott GW. Photochemistry and Photophysics. Volume II.1990.

22.

Omer A. Stydy on synthesis of coumarin derivatives. PhD Thesis. University of Mosul; 2001.

23.

Tanaka K. Supramolecular photodimerization of coumarins. Molecules. 2012;17:140818.

24.

Girotti R, Marrocchi A, Minuti L, Piermatti O, Pizzo F, Vaccaro L. Diels−Alder reactions of 3-substituted coumarins in water and under high-pressure condition. An uncatalyzed route to tetrahydro-6H-benzo[c]chromen-6-ones. Journal of Organic Chemistry. 2006;71(1):70–4.

25.

Bhardwaj Y, Chauhan M, Pareek A, Kishore D. Comparative study of the synthesis of 7hydroxy-4-methyl coumarin Mannich bases by conventional and microwave method. International Research Journal of Pure & Applied Chemistry. 2013;3(3): 250-6.

110

26.

Cooke D. Studies on the mode of action of coumarins (Coumarin, 6-hydroxycoumarin, 7hydroxycoumarin & Esculetin) at a cellular level. PhD Thesis. Dublin City University; 1999.

27.

Guilet D, Seraphin D, Rondeau D, Richomme P, Bruneton J. Cytotoxic coumarins from Calophyllum dispar. Phytochemistry. 2001;58:571-5.

28.

Keating G. Biosensor-based studies on coumarins. PhD Thesis. Dublin City University; 1998.

29.

Bogan D, Deasy B, O’Kennedy R, Smyth M. Determination of free and total 7hydroxycoumarin in urine and serum by capillary electrophoresis. J Chromatogr B. 1995;663:371-8.

30.

Cacini W, Ritschel WA. The excretion of coumarin and hydroxycoumarins by the avian kidney in vivo. Arch Int Pharmacodyn Ther. 1980;243(2):197-208.

31.

Pérez-Rodríguez E, Aguilera J, Gómez I, Figueroa F. Excretion of coumarins by the mediterranean green alga Dasycladus vermicularis in response to environmental stress. Marine Biology. 2001;139(4):633-9.

32.

Aguilar F, Autrup HN, Barlow S, Castle L, Crebelli R, Dekant W, et al. Coumarin in flavourings and other food ingredients with flavouring properties. European Food Safety Authority. 2008;793:1-15.

33.

Shi J, Li C-J, Yang J-Z, Ma J, Wang C, Tang J, et al. Hepatoprotective coumarins and secoiridoids from Hydrangea paniculata. Fitoterapia. 2014;96:138–45.

34.

Sproll C, Ruge W, Andlauer C, Godelmann R, Lachenmeier DW. HPLC analysis and safety assessment of coumarin in foods. Food Chemistry. 2008;109:462–9.

35.

Lee J-H, Kim Y-G, Cho H-S, Ryu S-Y, Cho M-H, Lee J. Coumarins reduce biofilm formation and the virulence of Escherichia coli O157:H7. Phytomedicine. 2014;21:1037– 42.

36.

Pan T-L, Wang P-W, Aljuffali IA, Leu Y-L, Hung Y-Y, Fang J-Y. Coumarin derivatives, but not coumarin itself, cause skin irritation via topical delivery. Toxicology Letters. 2014;226:173–81.

37.

Pangal A, Gazge M, Mane V, Shaikh JA. Various pharmacological aspects of couamrin derivatives: A review. International Journal Of Pharmaceutical Research And BioScience. 2013;2(6):168-94. 111

38.

Alipour M, Khoobi M, Emami S, Fallah-benakohal S, Ghasemi-Niri SF, Abdollahi M, et al. Antinociceptive properties of new coumarin derivatives bearing substituted 3,4dihydro-2H-benzothiazines. DARU Journal of Pharmaceutical Sciences. 2014;22:9.

39.

Behrami A, Krasniqi I, Demaku S. Antibacterial activity of coumarine derivatives synthesized from 4,7-dihydroxy-2-oxo-2H-chromene-3-carbaldehyde and comparison with standard drug. International Journal of Research in Pharmaceutical and Biomedical Sciences. 2012;3(3):1306-11.

40.

Cho J-Y, Hwang T-L, Chang T-H, Lim Y-P, Sung P-J, Lee T-H, et al. New coumarins and anti-inflammatory constituents from Zanthoxylum avicennae. Food Chemistry. 2012;135:17–23.

41.

Nath P, Dhumwad SD. Synthesis, characterization and antimicrobial studies of Co(II), Ni(II), Cu(II) and Zn(II) complexes derived from a Schiff base of 2-[(4-Methyl-2-oxo2Hchromen-7-yl)oxy]acetohydrazide with 3-formyl-2-hydroxy quinoline and 3-formyl-2mercapto quinoline. Journal of Chemical and Pharmaceutical Research. 2012;4(1):85165.

42.

Singh I, Kaur H, Kumar S, Kumar A, Lata S, Kumar A. Synthesis of new coumarin derivatives as antibacterial agents. International Journal of ChemTech Research. 2010;2:1745-52.

43.

Swamy MJ, Nandini GM, Pramod N. Synthesis, characterization and antifungal activity of coumarin carbohydrazide containing quinoline derivatives. International Journal of Research in Pharmaceutical and Nano Sciences. 2013;2(2):195- 202.

44.

Aly ES, Fodah ER, Saleh SY. Antiobesity, antioxidant and cytotoxicity activities of newly synthesized chalcone derivatives and their metal complexes. European Journal of Medicinal Chemistry. 2014;76:517-30.

45.

Čačić M, Pavić V, Molnar M, Šarkanj B, Has-Schon E. Design and synthesis of some new 1,3,4-thiadiazines with coumarin moieties and their antioxidative and antifungal activity. Molecules. 2014;19:1163-77.

46.

Chih-Chia C, Ling-Yih H, Hou-Jen T, Chen-Wen Y, Tsu-Chung C. Synthesis and antimicrobial evaluation of coumarin derivatives. Journal of CCIT. 2008;37(1):15-22.

47.

Hasanen JA. Synthesis and mass spectra of some new 3-substituted coumarin derivatives. Der Pharma Chemica. 2012;4(5):1923-34. 112

48.

Ibrahim M, Al–Sabea N, Saour K. Synthesis of nitrocoumarin derivative and separation of its isomers. AJPS. 2005;1(2):52-62.

49.

Lin P-Y, Yeh K-S, Su C-L, Sheu S-Y, Chen T, Ou K-L, et al. Synthesis and antibacterial activities of novel 4-hydroxy-7-hydroxy- and 3-carboxycoumarin derivatives. Molecules. 2012;17:10846-63.

50.

Sandhu S, Bansal Y, Silakari O, Bansal G. Coumarin hybrids as novel therapeutic agents. Bioorganic & Medicinal Chemistry. 2014;22:3806–14.

51.

Srivastav VK, Tiwari M. QSAR and docking studies of coumarin derivatives as potent HIV-1 integrase inhibitors. Arabian Journal of Chemistry. 2013;1(15):1-14.

52.

Miglietta A, Bocca C, Gabriel L, Rampa A, Bisi A, Valenti P. Antimicrotubular and cytotoxic activity of geiparvarin analogues, alone and in combination with paclitaxel. Cell Biochem Funct. 2001;19(3):181-9.

53.

Chen Y, Liu H-R, Liu H-S, Cheng M, Xia P, Qian K, et al. Antitumor agents 292. Design, synthesis and pharmacological study of S- and O-substituted 7-mercapto- or hydroxy-coumarins and chromones as potent cytotoxic agents. European Journal of Medicinal Chemistry. 2012;49:74-85.

54.

Forejtn´ıkov´a H, Lunerov´a K, Kub´ınov´a R, Jankovsk´a D, Marek R, Kareˇs R, et al. Chemoprotective and toxic potentials of synthetic and natural chalcones and dihydrochalcones in vitro. Toxicology. 2005;208:81–93.

55.

Piotrowska DG, Cieslak M, Królewska K, Wróblewski AE. Design, synthesis and cytotoxicity of a new series of isoxazolidines derived from substituted chalcones. European Journal of Medicinal Chemistry. 2011;46:1382-9.

56.

Sashidhara KV, Kumar A, Kumar M, Sarkar J, Sinha S. Synthesis and in vitro evaluation of novel coumarin–chalcone hybrids as potential anticancer agents. Bioorganic & Medicinal Chemistry Letters. 2010;20:7205–11.

57.

Syam S, Abdelwahab SI, Al-Mamary MA, Mohan S. Synthesis of chalcones with anticancer activities. Molecules. 2012;17:6179-95.

58.

Pingaew R, Prachayasittikul S, Ruchirawat S. Synthesis, cytotoxic and antimalarial activities of benzoyl thiosemicarbazone analogs of isoquinoline and related compounds. Molecules. 2010;15:988-96.

113

59.

Gomha SM, Khalil KD. A convenient ultrasound-promoted synthesis of some new thiazole derivatives bearing a coumarin nucleus and their cytotoxic activity. Molecules. 2012;17:9335-47.

60.

Mashelkar UC, Audi AA. Synthesis of some novel 4-substituted coumarins having potential biological activity. Indian Journal of Chemistry. 2006;45:1463-9.

61.

Mosa AI, Emara A, Yousef JM, Saddiq A. Novel transition metal complexes of 4hydroxy-coumarin-3-thiocarbohydrazone: Pharmacodynamic of Co(III) on rats and antimicrobial activity. Spectrochimica Acta. 2011;81:35– 43.

62.

Darla MM, Krishna BS, Rao KU, Reddy NB, Srivash MK, Adeppa K, et al. Synthesis and bio-evaluation of novel 7-hydroxy coumarin derivatives via Knoevenagel reaction. Res Chem Intermed. 2013.

63.

He X, Chen Y-Y, Shi J-B, Tang W-J, Pan Z-X, Dong Z-Q, et al. New coumarin derivatives: Design, synthesis and use as inhibitors of hMAO. Bioorganic & Medicinal Chemistry. 2014;22:3732–8.

64.

Kadhum AH, Al-Amiery AA, Musa AY, Mohamad AB. The antioxidant activity of new coumarin derivatives. International Journal of Molecular Sciences. 2011;12:5747-61.

65.

Lin H-C, Tsai S-H, Chen C-S, Chang Y-C, Lee C-M, Lai Z-Y, et al. Structure–activity relationship of coumarin derivatives on xanthine oxidase-inhibiting and free radicalscavenging activities. Biochemical pharmacology. 2008;75:1416 – 25.

66.

Sashidhara KV, Palnati GR, Sonkar R, Avula SR, Awasthi C, Bhatia G. Coumarin chalcone fibrates: A new structural class of lipid lowering agents. European Journal of Medicinal Chemistry. 2013;64:422-31.

67.

Basile A, Sorbo S, Spadaro V, Bruno M, Maggio A, Faraone N, et al. Antimicrobial and antioxidant activities of coumarins from the roots of Ferulago campestris (Apiaceae). Molecules. 2009;14:939-52.

68.

Bubols G, Vianna D, Medina-Remón A, Poser Gv, Lamuela-Raventos RM, Eifler-Lima VL, et al. The antioxidant activity of coumarins and flavonoids. Mini-Reviews in Medicinal Chemistry. 2013;13(3):1-17.

69.

Choudhary S, Kini S, Mubeen M. Antioxidant activity of novel coumarin substituted benzothiazole derivatives. Der Pharma Chemica. 2013;5(4):213-22.

114

70.

Sandeep G, Ranganath YS, Bhasker S, Rajkumar N. Synthesis and biological screening of some novel coumarin derivatives. Asian J Research Chem. 2009;2(1):46-8.

71.

Arshad A, Osman H, Bagley MC, Lam CK, Mohamad S, Zahariluddin ASM. Synthesis and antimicrobial properties of some new thiazolyl coumarin derivatives. European Journal of Medicinal Chemistry. 2011;46:3788-94.

72.

Basanagouda M, Jambagi VB, Barigidad NN, Laxmeshwar SS, Devaru V, Narayanachar. Synthesis, structureeactivity relationship of iodinated-4-aryloxymethyl-coumarins as potential anti-cancer and antimycobacterial agents. European Journal of Medicinal Chemistry. 2014;74:225-33.

73.

Kostova I, Nikolova I. Stability of Neodymium(iii) complexes of 4-hydroxycoumarins with anticoagulant activity. American Journal of Pharmacology and Toxicology. 2006;1(2):30-5.

74.

Aslam K, Khosa MK, Jahan N, Nosheen S. Synthesis and applications of coumarin. Pak J Pharm Sci. 2010;23(4):449-54.

75.

Sashidhara KV, Kumar A, Kumar M, Sonkar R, Bhatia G, Khanna AK. Novel coumarin derivatives as potential antidyslipidemic agents. Bioorganic & Medicinal Chemistry Letters. 2010;20:4248–51.

76.

Beyhan N, Kocyigit-Kaymakcioglu B, Gumru S, Aricioglu F. Synthesis and anticonvulsant activity of some 2-pyrazolines derived from chalcones. Arabian Journal of Chemistry. 2013;7(37):1-9.

77.

Mantri N, Jadon G. To Synthesized Mannich bases of 7-hydroxy-4-methyl coumarin with different secondary amine. International Journal of Advanced Research in Pharmaceutical And Bio Sciences. 2013;3(2):117-26.

78.

Lamani KS, Kotresh O, Phaniband MA, Kadakol JC. Synthesis, characterization and antimicrobial properties of Schiff bases derived from condensation of 8-Formyl-7hydroxy-4-methylcoumarin and substituted triazole derivatives. E-Journal of Chemistry. 2009;6:239-46.

79.

Halli MB, Sumathi RB, Kinni M. Synthesis, spectroscopic characterization and biological evaluation studies of Schiff’s base derived from naphthofuran-2-carbohydrazide with 8formyl-7-hydroxy-4-methyl coumarin and its metal complexes. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy. 2012;99:46–56. 115

80.

Manidhar DM, Rao UM, Reddy NB, Sundar CS, Reddy CS. Synthesis of new 8-formyl4-methyl-7-hydroxy coumarin derivatives. Journal of the Korean Chemical Society. 2012;56(4):459-63.

81.

Dömötör O, Tuccinardi T, Karcz D, Walsh M, Creaven BS, Enyedy ÉA. Interaction of anticancer reduced Schiff base coumarin derivatives with human serum albumin investigated by fluorescence quenching and molecular modeling. Bioorganic Chemistry. 2014;52:16–23.

82.

Kulkarni A, Patil SA, Badami PS. Synthesis, characterization, DNA cleavage and in vitro antimicrobial studies of La(III), Th(IV) and VO(IV) complexes with Schiff bases of coumarin derivatives. European Journal of Medicinal Chemistry. 2009;44:2904–12.

83.

Pangal A, Shaikh JA, Gazge M, Mane V, Ahmed K. Novel 3-acetylcoumarinSchiff's base synthesis from different acid hydrazide. International Research Journal of Pharmacy. 2013;4(10):108-10.

84.

Ali RT. Synthesis of Schiff bases derived from benzaldehyde and salicylaldehyde with some amino acids by a new develop method. National Journal of Chemistry. 2010;37:158-68.

85.

Kumar DN, Sharma P, pareek A. Synthesis of new Schiff base complexes and their applications. International Journal of Applied Research & Studies. 2013;2:307.

86.

Rîmbu C, Danac R, Pui A. Antibacterial activity of Pd(II) complexes with salicylaldehyde amino acids Schiff bases ligands. Chem Pharm Bull. 2014;62:12-5.

87.

Mielcke TR, Mascarello A, Filippi-Chiela E, Zanin RF, Lenz G, Leal PC, et al. Activity of novel quinoxaline-derived chalcones on in vitro glioma cell proliferation. European Journal of Medicinal Chemistry. 2012;48:255-64.

88.

Smit FJ, N’Da D. Synthesis, in vitro antimalarial activity and cytotoxicity of novel 4aminoquinolinyl-chalcone amides. Bioorganic & Medicinal Chemistry. 2014;22:1128– 38.

89.

Zuo Y, Yu Y, Wang S, Shao W, Zhou B, Lin L, et al. Synthesis and cytotoxicity evaluation of biaryl-based chalcones and their potential in TNFa-induced nuclear factorkB activation inhibition. European Journal of Medicinal Chemistry. 2012;50:393-404.

90.

Song Z-K, Dong B, Lei G-J, Peng M-J, Guo Y. Novel selective fluorescent probes for sensing Zn2+ ions based on a coumarin Schiff base. Tetrahedron Letters. 2013;54:4945–9. 116

91.

Serda M, Musiol R, Polanski J. New thiosemicarbazones based on quinoline scaffold as anticancer iron chelators. 14th International Electronic Conference on Synthetic Organic Chemistry (ECSOC-14). 2010.

92.

Mohamed HM, El-Wahab FA, Ahmed KA, El-Agrody AM, Bedair AH, Eid FA, et al. Synthesis, reactions and antimicrobial activities of 8-ethoxycoumarin derivatives. Molecules. 2012;17:971-88.

93.

Al-Kawkabani A, Boutemeur-Kheddis B, Makhloufi-Chebli M, Hamdi M, Talhi O, Silva MS. Synthesis of novel 2H,8H-pyrano[2,3-f]chromene-2,8-diones from 8-formyl-7hydroxy-4-methylcoumarin. Tetrahedron Letters. 2013;54:5111–4.

94.

Department of Chemistry DGC, Darjeeling, West Bengal, India. Generalized valence bond

orbital

interactions

(GVB-OIs)

and

Huckel’s

4n

+

2/4n

rule

for

aromaticity/antiaromaticity. Journal of Molecular Structure (Theochem). 1995;337:151-4. 95.

Figeys HP. Quantum-chemical studies on the aromaticity of conjugated systems-II aromatic and anti-aromatic annulenes: The (4n + 2)-p electron rule. Tetrahedron. 1970;26:5225-34.

96.

Smithetrman HC, Ferguson LN. On the aromatic character of 4-pyrones. Tetrahedron. 1968;24:923-32.

97.

Balaji P, Seleena S, Vidya G, Himateja T, Neelima B, Lakshmiteja T. Synthesis and charecterisation of Schiff bases derived from acetyl coumarin and evaluated for antimicrobial activity. Research Journal of Pharmaceutical, Biological and Chemical Sciences. 2013;4(2):9.

98.

Pingaew R, Saekee A, Mandi P, Nantasenamat C, Prachayasittikul S, Ruchirawat S, et al. Synthesis, biological evaluation and molecular docking of novel chalcone-coumarin hybrids as anticancer and antimalarial agents. European Journal of Medicinal Chemistry. 2014;85:65-76.

99.

Klančnik A, Piskernik S, Jeršek B, Možina S. Evaluation of diffusion and dilution methods to determine the antibacterial activity of plant extracts. Journal of Microbiological Methods. 2010;81:121–6.

100.

Nagayama A, Yamaguchi K, Watanabe K, Tanaka M, Kobayashi I, Nagasawa Z. Final report from the committee on antimicrobial susceptibility testing. Japanese Society of Chemotherapy. 2008;14:383–92. 117

101.

Tille PM. Bailey & Scott's Diagnostic Microbiology.Thirteenth edition. 2014.

102.

Patrick MR. Manual of Clinical Microbiology. Sixth edition. 2005.

103.

Cetin-Karaca H. Evaluation of natural antimicrobial phenolic compounds against foodborne pathogens. MSc Thesis. University of Kentucky; 2011.

104.

Ferna´ndez-Torres B, Inza I, Guarro J. Evaluation of disk diffusion method for determining Eberconazole susceptibility of dermatophytes and influence of culture medium. Antimicrobial agents and Chemotherapy. 2005;49(5):2116–8.

105.

Greenwood D, Barer M, Slack R, Irving W. Medical Microbiology. Eighteenth edition. 2012.

118

shokhan- MSc thesis pdf.pdf

PHARMACEUTICAL CHEMISTRY. By. Shokhan Jamal Hamid. B.Sc. Pharmacy 2010. Supervised by. Dr. Ammar A. Mahmood Kubba. PhD Pharmaceutical ...

4MB Sizes 7 Downloads 307 Views

Recommend Documents

shokhan- MSc thesis pdf.pdf
Pharmacognosy & Pharmaceutical Chemistry for his tremendous help, support and. encouragement. Words fail to express my humble gratitude to Dr. Emad ...

Msc Thesis Yousif.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. Msc Thesis ...

David Burg - MSc Thesis
1.1.5 Nuclear Transport, Integration and Gene Expression . ...... (1997) analyzed data from experimental infection of macaques with SIVsm ... buffered solution containing DNA polymerase, oligonucleotide primers, the four deoxynucleotide.

MSc thesis - Center for Development Research (ZEF)
Being enrolled in a masters program on agriculture, forestry, geography, intermingled or related discipline. • Proficiency in biophysical and socioeconomic field ...

David Burg - MSc Thesis
Oct 2, 2010 - dynamics is a powerful tool in research of viral dynamics, has changed ..... humans, and using analytical tools devised by Nowak et al., thereby ...

MSC thesis-Effect of Some Medicinal Drugs.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item.

My MSc Thesis - Yadgar Ibrahim Abdulkarim.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. My MSc Thesis ...

MSC-Thesis-Parween-PDF - parween Othman Qader.pdf ...
There was a problem loading this page. MSC-Thesis-Parween-PDF - parween Othman Qader.pdf. MSC-Thesis-Parween-PDF - parween Othman Qader.pdf.

Shokhan and Shanaz.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. Shokhan and ...Missing:

MSC SINFONIA
Mar 18, 2015 - R 6 200. R 3 100. Ocean View Cabin. 7 000. 3 500. Balcony Cabin ..... lking Track. Power Wa lking Track. Pasha Club Disco. Galaxy. Video.

Dr. Shokhan Rasool Ahmed3.pdf
Page 3 of 126. WITCHCR AFT AT COURT AND THE GLOBE. Shokhan Ahmed. THE STAGING OF WITCHCRAFT. and a. “SPECTACLE OF STRANGENESS”. Page 3 of 126. Dr. Shokhan Rasool Ahmed3.pdf. Dr. Shokhan Rasool Ahmed3.pdf. Open. Extract. Open with. Sign In. Main m

Dr. Shokhan Rasool Ahmed Magic1.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. Dr. Shokhan ...

MSc Computer Science.pdf
MSc Computer Science.pdf. MSc Computer Science.pdf. Open. Extract. Open with. Sign In. Main menu. Displaying MSc Computer Science.pdf.

Bachelor Thesis - arXiv
Jun 26, 2012 - system such as Solr or Xapian and to design a generic bridge ..... application server. ..... document types including HTML, PHP and PDF.

Bachelor Thesis - arXiv
Jun 26, 2012 - Engine. Keywords. Document management, ranking, search, information ... Invenio is a comprehensive web-based free digital library software.

Master's Thesis - CiteSeerX
Some development activist, on the other hand, considered the ... Key-words: Swidden agriculture; Chepang; land-use change; environmental perception ...

Master's Thesis - Semantic Scholar
... or by any means shall not be allowed without my written permission. Signature ... Potential applications for this research include mobile phones, audio production ...... [28] L.R. Rabiner and B. Gold, Theory and application of digital signal ...

Thesis Proposal.pdf
Architect : Rem Koolhaas. Location : Utrecht , Holland. Area : 11,000 Sq.m. Completion : 1998. EDUCATORIUM. Utrecht University , Holland. Page 4 of 23.

Master Thesis - GitHub
Jul 6, 2017 - Furthermore, when applying random initialization, we could say a “warmup” period is required since all ..... that is, the worker will move back towards the central variable. Nevertheless, let us ... workers are not able to move, eve

Master's Thesis - CiteSeerX
Aug 30, 2011 - purposes, ranging from grit of maize as substitute of rice, for making porridge, local fermented beverage, and fodder for poultry and livestock. In both areas the fallow period however has been reduced from 5-10 years previously to 2-4

Tsetsos thesis
Mar 15, 2012 - hand, value-based or preferential choices, such as when deciding which laptop to buy ..... nism by applying small perturbations to the evidence and showing a larger .... of evidence integration these two models would be equally good ..

thesis-submitted.pdf
Professor of Computer Science and. Electrical and Computer Engineering. Carnegie Mellon University. Page 3 of 123. thesis-submitted.pdf. thesis-submitted.pdf.

Master's Thesis - CiteSeerX
Changes in major land-use(s) in Jogimara and Shaktikhar between ...... Angelsen, A., Larsen, H.O., Lund, J.F., Smith-Hall, C. and Wunder, S. (eds). 2011.