+ All Categories
Home > Documents > prr.hec.gov.pkprr.hec.gov.pk/jspui/bitstream/123456789/2521/1/2577S.pdf · i ACKNOWLEDGEMENTS The...

prr.hec.gov.pkprr.hec.gov.pk/jspui/bitstream/123456789/2521/1/2577S.pdf · i ACKNOWLEDGEMENTS The...

Date post: 25-Oct-2019
Category:
Upload: others
View: 10 times
Download: 0 times
Share this document with a friend
180
SYNTHESIS, CHARACTERIZATION AND BIOASSAY OF SOME NOVEL 6,8-DIOXYGENATED-7-SUBSTITUTED ISOCOUMARINS, DIHYDROISOCOUMARINS AND RELATED COMPOUNDS Islamabad A dissertation submitted to the Department of Chemistry, Quaid-i-Azam University, Islamabad, in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Organic Chemistry by Muhammad Zaman Ashraf Department of Chemistry Quaid-i-Azam University Islamabad 2010
Transcript

SYNTHESIS, CHARACTERIZATION AND BIOASSAY OF SOME

NOVEL 6,8-DIOXYGENATED-7-SUBSTITUTED ISOCOUMARINS,

DIHYDROISOCOUMARINS AND RELATED COMPOUNDS

Islamabad

A dissertation submitted to the Department of Chemistry,

Quaid-i-Azam University, Islamabad, in partial fulfillment

of the requirements for the degree of

Doctor of Philosophy

in

Organic Chemistry

by

Muhammad Zaman Ashraf

Department of Chemistry

Quaid-i-Azam University

Islamabad

2010

To

MY MOTHER

AND

MY WIFE SAMMAR ZAMAN

i

ACKNOWLEDGEMENTS

The entire gamut of creation amplify figures out, in all its dimensions, latent or

manifest, the intricately balanced interplay of its fundamental determinants – knowledge

and practice – which while ordering the elementary constituents, quite distinctly

identifiable in their own right, impacts the whole a fascinating and majestic character that

doubtless evokes the intuitive response of gratitude and indebtedness to the Immanent,

the Transcendent, the Omnipotent – ALLAH.

The human drive to fashion the evolving universal framework in a peculiarly

human perspective, our beloved Prophet MUHAMMD (Peace and Blessing of Allah

be upon him and his descendents) stands for an ever actuating and galvanizing

impulsive force that brings out the divine through the human. Indeed He commands the

highest order amongst the whole creation.

I consider it my first and foremost obligation to express my heartful gratitude to

my respected and inspirational supervisor, Dr. Aamer Saeed. I would not have been able

to complete this assignment without his cordial attitude, kidness, guidance and persistent

interest in my research work.

I am thankful to Chairman Department of Chemistry, Professor Dr. Saqib Ali and

Head of Organic Section, Professor Dr. Nasim Hasan Rama for providing me basic

research facilities. I am thankful to all faculty members, department of chemistry Quaid-

i-Azam University, Islamabad.

I am grateful to Prof. Dr. Peter Langer Rostock, Germany, for cytotoxic studies

of my samples. I am thankful to Dr. Muhammad Ali, HEJ institute, Karachi, for

antimalarial activity and Dr. Imran Arid Agriculture University, Rawalpindi for

antimicrobial assay of my synthesized compounds.

I am thankful to all my lab fellows Ammara Mumtaz, Naeem Abbass, Hummera

Rafique, Rasheed Ahmed Kherra, Madiha Irfan, Aliya Ibrar, Uzma Shaheen, Iram Batool

and Shumaila Bukhari for their cooperation and support. I would like to acknowledge my

friends Shahid Ameen Samra, Latif Hussain, Dr. Muhammad Sher, Latif Shahid, Latif

Shahid, Muhammd Waqas, Yasir Asghar, Safdar sb. Kamran, Imran, my colleagues,

ii

Prof. Dr. Saeed, Prof. Dr. Karamat Ali, Prof. Dr. G.A. Miana, Dr. Naghmana Rashid,

Dr. Uzma Yunas, Dr. Imtiaz, Dr. Moazzam, Ziaullah Shah, Aun Muhammad, Tariq

Ismail, Khurram Afzal, Muhammad Imran, Dr. Humaira Nadeem, Miss Kishwar and

Najm-ul-Hassan. I am thankful to all my students at RIPS for their prayers.

I have no words to acknowledge the prayers, support, encouragement and

dedication of my loving parents as the whole I achieved is actually their achievement and

they certainly deserve more than this along with my sister and loving brothers. Their

prayers and encouragement were always with me till the completion of my thesis. My

special love is for my wife who always encouraged and assisted me in completion of this

project along with my adorable kids Hishma Zaman and Muhammad Taha Zaman.

I want to acknowledge my loving Baji Nasim, Baji Shameem, Baji Nasreen,

Mudassir, Ammi and Abba ji who always remember me in their prayers.

At the end I am thankful to all those hands that prayed for my betterment and

success.

(Muhammad Zaman Ashraf)

iii

Abstract

The work presented in this thesis has been divided into two parts. Part one deals

with the synthesis, characterization and biological activity of some 7-substituted 6,8-

dioxygenated isocoumarins and 3,4-dihydroisocoumarins. Chapter one includes general

introduction, nomenclature, structural types, biosynthesis, and extensive examples on

pharmacological efficacy of isocoumarins and 3,4-dihydroisocoumarins from literature. It

also provides some of the most significant synthetic routes and the reactions of

isocoumarins and 3,4-dihydroisocoumarins and their interconversion.

The total synthesis of structural analogues of some naturally occurring bioactive

isocoumarins and dihydroisocoumarins viz. Hiburipyranone, Cytogenin, Montroumarin,

Scorzocreticin, Annulatomarin, Thunberginol B, starting from 3,5-dimethoxy-4-methyl

homophthalic acid is the subject of Chapter two. The synthesis of 3,5-dimethoxy-4-

methylhomophthalic acid from simplest precursor p-toluic acid was carried out. The

substituted homophthalic acid was then converted into corresponding anhydride which

was then condensed with various acyl and aroyl chlorides to afford the corresponding 3-

alkyl or 3-arylisocoumarins. The isocoumarins were then converted into corresponding

3,4-dihydroisocoumarins and the latter were then demethylated to afford corresponding

6,8-dihydroxy-3,4-dihydroisocoumarins. The structures of all of the synthesized

compounds were confirmed using FTIR, 1H NMR,

13C NMR and mass spectral data.

Chapter three provides the physical constants and spectroscopic data of the synthesized

compounds.

Chapter four deals with the biological activities of the compounds synthesized.

Antibacterial activity was determined against ten different Gram positive and Gram

negative bacterial strains (Micrococcus luteus, Staphylococcus aureus, Staphylococcus

epidermidis, Lactobacillus bulgaricus, Escherichia coli, Klebsiella pneumonae,

Pasteurella multocida, Proteus vulgaris, Pseudomonas aeruginosa and Salmonella typhi)

using agar well diffusion method. In vitro antimalarial activity was performed against

malarial parasite Plasmodium falciparum. The cytotoxic activity of the synthesized

compounds was determined against human keratinocyte cell lines.

Chapter five depicts total synthesis of a natural product 8-hydroxy-7-

hydoxymethyl-6-methoxy-3,4-dihydroisocoumarin (Stellatin) isolated from mycelium of

iv

Aspergillus variecolor. The structures of the precursor compounds and the Stellatin were

determined by FTIR, NMR and mass spectroscopic data. These compounds were

evaluated for their antibacterial activity against ten different gram positive and gram

negative bacterial strains. The cytotoxic activity was performed against human

keratinocyte cell lines.

Part two is related to the synthesis of some 3-(substituted phenyl)isocoumarins, 3-

(substituted phenyl)isocoumarin-1-thiones, 3-(substituted phenyl)isoquinolones and some

1-aryl-7,8-dichloroisochromans. Chapter seven, after general introduction, describes the

synthesis and biological activity of these compounds. The unsubstituted homophthalic

acid was converted into anhydride by treatment with acetic anhydride. The latter was then

converted into 3-(substituted phenyl)isocoumarins by reacting it with suitable acid

chlorides. The isocoumarins were then converted into corresponding 3-(substituted

phenyl)isoquinolones by treatment with formamide. The 3-(substituted phenyl)

isocoumarin-1-thiones were synthesized from isocoumarins using Lawesson’s reagent

under microwave irradiation. Microwave assisted synthesis of some (±)-1-aryl-7,8-

dichloroisochromans was carried out by condensation of 2-(3,4-dimethoxyphenyl)

ethanol with a variety of aromatic aldehydes via an acid catalyzed oxa-Pictet-Spengler

reaction.

All of these synthesized compounds were characterized by IR, 1H,

13C NMR and

mass spectroscopic data. In vitro antibacterial activity of these compounds was

determined against ten different Gram positive and Gram negative bacterial strains using

agar well diffusion method.

The comparative analysis of the antibacterial activity of the 3-(substituted

phenyl)isocoumarins, 3-(substituted phenyl)isocoumarin-1-thiones and 3-(substituted

phenyl)isoquinolones is described. Accordingly, the antibacterial activity increases when

isocoumarins were converted into corresponding isocoumarin-1-thiones but decreases on

conversion into corresponding isoquinolones.

v

Table of Contents

Acknowledgement i-ii

Abstract iii-iv

Contents v-xvi

PART I 1-125

CHAPTER ONE 1-35

Introduction

1.1 Nomenclature and Structural Type 1

1.2 Physical Properties 4

1.3 Biosynthesis 4

1.4 Pharmacological Applications 11

1.5 Synthesis of Isocoumarins and 3,4-Dihydroisocoumarins 17

1.5.1 Oxidation of Isochromans 17

1.5.2 Oxidation of Indenes, Indanones and Indones 18

1.5.3 Synthesis involving Metals 20

a) Lithiation 20

b) Thallation-olefination of Arenes 22

c) Silylation 22

d) Organo-mercury catalyzed synthesis 23

e) Palladium catalyzed method 24

f) Iridium catalyzed method 24

g) Rhodium-Catalyzed Oxidative Coupling of Benzoic Acids with

Alkynes via Regioselective C-H Bond Cleavage 25

1.5.4 Aldol-type Condensation between Homophthalic Acids,

Esters or Anhydrides and Carbonyl Compounds 25

a) Stobbe Condensation of Homophthalates with

Aldehydes and Ketones 26

b) Claisen Condensation of Homophthalates with Formates 26

c) Claisen Condensations of Homophthalates with Oxalates 27

d) Condensation of Malonyl Heterocycles with diphenylcarbonate 28

vi

e) Condensation of Acid chlorides, Phenols,

Phenol Acids with Homophthalic Acids and Anhydrides 28

1.6 Reactions of Isocoumarins and 3,4-dihydroisocoumarins 30

1.6.1 Hydrolysis 30

1.6.2 Reaction with Ammonia and Amines 31

1.6.3 Reaction with Phosphorus Pentasulfide 32

1.6.4 Nitration 32

1.6.5 Reaction with Grignard Reagents 33

1.6.6 Oxidation 33

1.6.7 Reduction 33

1.7 Interconverision of Isocoumarins and 3,4-Dihydroisocumarins 33

1.7.1 Conversion of 3,4-Dihydroisocoumarins to Isocoumarins 34

a) Alkaline Hydrolysis Followed by Oxidation and Recyclization 34

b) Benzylic Bromination Followed by Dehydrobromination 34

1.7.2 Conversion of Isocoumarins to 3,4-Dihydroisocoumarins 34

a) Alkaline Hydrolysis Followed by Reduction and Recyclization 34

b) Catalytic Reduction 35

CHAPTER TWO 36-55

Experimental

2.1 Purification of Solvents 36

2.2 Instrumentation 36

2.3 Synthesis of methyl 4-methylbenzoate (1) 36

2.4 Synthesis of 3,5-dibromo-4-methylbenzoate (2) 36

2.5 3,5-Dimethoxy-4-methyl benzoic acid (3) 37

2.6 Methyl 3,5-dimethoxy-4-methyl benzoate (4) 37

2.7 (3,5-Dimethoxy-4-methyl phenyl)methanol (5) 37

2.8 3,5-Dimethoxy-4-methyl benzyl bromide (6) 38

2.9 (3,5-Dimethoxy-4-methylphenyl) acetonitril (7) 38

2.10 (3,5-Dimethoxy-4-methyl phenyl) acetic acid (8) 38

2.11 Methyl (3,5-dimethoxy-4-methyl phenyl) acetate (9) 38

2.12 Methyl (2-formyl-3,5-dimethoxy-4-methyl phenyl) acetate (10) 39

vii

2.13 2,4-Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid(11) 39

2.14 6-(Carboxymethyl)-2,4-dimethoxy-3-methylbenzoic acid (12) 40

2.15 6,8-Dimethoxy-7-methyl-1H-isochromene-1,3(4H)-dione (13) 40

2.16 General procedure for 6,8-dimethoxy-7-methyl-3-alkyl/arylisocoumarins

(16a-j) 41

2.17 3-Propyl-6,8-dimethoxy-7-methylisocoumarin (16a) 41

2.18 3-Pentyl-6,8-dimethoxy-7-methylisocoumarin (16b) 42

2.19 3-Heptyl-6,8-dimethoxy-7-methylisocoumarin (16c) 42

2.20 3-Chloromethyl-6,8-dimethoxy-7-methylisocoumarin (16d) 42

2.21 3-Hydroxymethyl-6,8-dimethoxy-7-methylisocoumarin (16e) 43

2.22 3-Phenyl-6,8-dimethoxy-7-methylisocoumarin (16f) 43

2.23 3-(2-Chlorophenyl)-6,8-dimethoxy-7-methylisocoumarin (16g) 43

2.24 3-(4-Methoxyphenyl)-6,8-dimethoxy-7-methylisocoumarin (16h) 43

2.25 3-(3,4-Dimethoxyphenyl)-6,8-dimethoxy-7-methylisocoumarin(16i)44

2.26 3-(3,4,5-Trimethoxyphenyl)-6,8-dimethoxy-7-methylisocoumarin (16j) 44

2.27 General procedure for 2,4-dimethoxy-3-methyl-6-(2-oxoalkyl/aryl)benzoic

acid (17a-j) 44

2.28 2,4-Dimethoxy-3-methyl-6-(2-oxopentyl)benzoic acid (17a) 45

2.29 2,4-Dimethoxy-3-methyl-6-(2-oxoheptyl)benzoic acid (17b) 45

2.30 2,4-Dimethoxy-3-methyl-6-(2-oxononyl)benzoic acid (17c) 45

2.31 6-(3-Chloro-2-oxopropyl)-2,4-dimethoxy-3-methylbenzoic

acid(17d) 45

2.32 6-(3-Hydroxy-2-oxopropyl)-2,4-dimethoxy-3-methylbenzoic

acid (17e) 46

2.33 2,4-Dimethoxy-3-methyl-6-(2-oxo-2-phenylethyl)benzoic

acid (17f) 46

2.34 6-[2-(2-Chlorophenyl)-2-oxoethyl]-2,4-dimethoxy-

-3-methylbenzoic acid (17g) 46

2.35 2,4-Dimethoxy-6-[2-(4-methoxyphenyl)-2-oxoethyl]

-3-methylbenzoic acid (17h) 47

viii

2.36 2,4-Dimethoxy-6-[2-(3,4-dimethoxyphenyl)-2-oxoethyl]-3-methylbenzoic

acid (17i) 47

2.37 2,4-dimethoxy-6-[2-(3,4,5-trimethoxyphenyl)-2-oxoethyl]

-3-methylbenzoic acid (17j) 47

2.38 General procedure for 6,8-dimethoxy-7-methyl-3-

alkyl/aryl-3,4- dihydroisocoumarins (18a-j) 48

2.39 6,8-Dimethoxy-7-methyl-3-propyl-3,4-dihydroisocoumarins (18a) 48

2.40 6,8-Dimethoxy-7-methyl-3-pentyl-3,4-dihydroisocoumarins (18b) 48

2.41 6,8-Dimethoxy-7-methyl-3-heptyl-3,4-dihydroisocoumarins (18c) 49

2.42 6,8-Dimethoxy-7-methyl-3-chloromethyl-3,4- 49

dihydroisocoumarins (18d)

2.43 6,8-Dimethoxy-7-methyl-3-hydroxymethyl-3,4- 49

dihydroisocoumarins (18e)

2.44 6,8-Dimethoxy-7-methyl-3-phenyl-3,4-dihydroisocoumarins (18f) 50

2.45 6,8-Dimethoxy-7-methyl-3-(2-chlorophenyl)-3,4-

dihydroisocoumarins (18g) 50

2.46 6,8-Dimethoxy-7-methyl-3-(4-methoxyphenyl)-3,4-

dihydroisocoumarins (18h) 50

2.47 6,8-Dimethoxy-7-methyl-3-(3,4-dimethoxyphenyl)-3,4-

dihydroisocoumarins (18i) 51

2.48 6,8-Dimethoxy-7-methyl-3-(3,4,5-trimethoxyphenyl)-3,4-

dihydroisocoumarins (18j) 51

2.49 General procedure for 6,8-dihydroxy-7-methyl-3-alkyl/aryl-3,4-

dihydroisocoumarins (19a-j) 51

2.50 6,8-Dihydroxy-7-methyl-3-propyl-3,4-dihydroisocoumarins (19a) 52

2.51 6,8-Dihydroxy-7-methyl-3-pentyl-3,4-dihydroisocoumarins (19b) 52

2.52 6,8-Dihydroxy-7-methyl-3-heptyl-3,4-dihydroisocoumarins (19c) 52

2.53 6,8-Dihydroxy-7-methyl-3-chloromethyl-3,4-

-dihydroisocoumarins (19d) 53

2.54 6,8-Dihydroxy-7-methyl-3-hydroxymethyl-3,4-

dihydroisocoumarins (19e) 53

ix

2.55 6,8-Dihydroxy-7-methyl-3-phenyl-3,4-dihydroisocoumarins (19f) 53

2.56 6,8-Dihydroxy-7-methyl-3-(2-chlorophenyl)-3,4- 53

dihydroisocoumarins (19g)

2.57 6,8-Dihydroxy-7-methyl-3-(4-methoxyphenyl)-3,4-

dihydroisocoumarins (19h) 54

2.58 6,8-Dihydroxy-7-methyl-3-(3,4-dimethoxyphenyl)-3,4-

dihydroisocoumarins 54

2.59 6,8-Dihydroxy-7-methyl-3-(3,4,5-trimethoxyphenyl)-3,4-

dihydroisocoumarins (19j) 54

CHAPTER THREE 56-87

Results and Discussion

3.1 Synthesis of 3,5-dimethoxy-4-methylhomophthalic acid (12) 56

3.2 Synthesis of 6,8-dimethoxy-7-methyl-3-

alkyl/aryl isocoumarins (16a-j) 65

3.3 Synthesis of 6,8-dimethoxy-7-methyl-3-alkyl/aryl-

3,4-dihydroisocoumarins (18a-j) 72

3.4 Synthesis of 6,8-dihydroxy-7-methyl-3-alkyl/aryl-

3,4-dihydroisocoumarins (19a-j) 82

CHAPTER FOUR 88-106

Biological Activities

4.1 Antibacterial Activity 88

4.2 Antimalarial Activity 94

4.3 Cytotoxicity 100

4.3.1 Cytotoxic Activity of the Isocoumarins (16a-j) 100

4.3.2 Cytotoxic Activity of the Keto Acids (17a-j) 102

4.3.3 Cytotoxic Activity of the 3,4-Dihydroisocoumarins (18a-j) 103

4.3.4 Cytotoxic Activity of the 6,8-Dihydroxy-

3,4-Dihydroisocoumarins (19a-j) 104

CHAPTER FIVE 107-118

5.1 Synthesis of Stellatin 107

x

5.1.1 Methyl (3, 5-dimethoxy-4-methyl phenyl) acetate (1) 107

5.1.2 Methyl (2-formyl-3,5-dimethoxy-4-methyl phenyl) acetate (2) 107

5.1.3 2,4-Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid (3) 108

5.1.4 2,4-dimethoxy-6-(2-hydroxyethyl)-3-methylbenzoic acid (4) 108

5.1.5 6,8-dimethoxy-7-methyl-3,4-dihydro-1H-isochromen-1-one (5) 109

5.1.6 6,8-dimethoxy-7-(bromomethyl)-3,4-dihydro-

1H-isochromen-1-one (6) 109

5.1.7 7-(hydroxymethyl)-6,8-dimethoxy-3,4-dihydro-

1H-isochromen-1-one (7) 110

5.1.8 8-hydroxy-7-(hydroxymethyl)-6-methoxy-

3,4-dihydro-1H-isochromen-1-one (8) 110

5.2 Results and Discussion 111

5.3 Antibacterial Activity 116

5.4 Cytotoxic Activity 118

CHAPTER SIX References Part I 119-125

Part II 126-162

CHAPTER SEVEN

7.1 Introduction 126

7.2 Experimental 127

Synthesis of homophthalic anhydride (1)

General procedure for 3-alkyl/arylisocoumarins (4a-j) 127

3-(3-Fluorophenyl)isocoumarin (4a) 128

3-(4-Fluorophenyl)isocoumarin (4b) 128

3-(2-Chlorobenzyl)isocoumarin (4c) 128

3-(2-Bromophenyl)isocoumarin (4d) 129

3-(3-Iodophenyl)isocoumarin (4e) 129

3-(2,4-Dichlorophenyl)isocoumarin (4f) 129

3-(2-Chloro-4-fluorophenyl)isocoumarin (4g) 129

3-(3-Nitrophenyl)isocoumarin (4h) 130

3-(2-Chloropyridyl)isocoumarin (4i) 130

xi

3-Pentadecylisocoumarin (4j) 130

General procedure for the conversion of isocoumarins

into 1(2H)-isoquinolones (5a–j) 131

3-(3-Fluorophenyl)isoquinolin-1(2H)-one (5a) 131

3-(4-Fluorophenyl)isoquinolin-1(2H)-one (5b) 131

3-(2-Chlorobenzyl)isoquinolin-1(2H)-one (5c) 131

3-(2-Bromophenyl)isoquinolin-1(2H)-one (5d) 132

3-(3-Iodophenyl)isoquinolin-1(2H)-one (5e) 132

3-(2,4-Dichlorophenyl)isoquinolin-1(2H)-one (5f) 132

3-(2-Chloro-4-fluorophenyl)isoquinolin-1(2H)-one (5g) 133

3-(3-Nitrophenyl)isoquinolin-1(2H)-one (5h) 133

3-(2-Chloropyridyl)isoquinolin-1(2H)-one (5i) 133

3-Pentadecylisoquinolin-1(2H)-one (5j) 134

7.3 Results and Discussion 134

Synthesis of 3-phenyl substituted-1H-isochromen

-1-thiones 138

7.4 Experimental 138

General procedure for the conversion of isocoumarins

into 1-1H-isochromene-1-thiones (2a–j) 138

3-(3-Fluorophenyl)-1H-isochromene-1-thione (7a) 138

3-(4-Fluorophenyl)-1H-isochromene-1-thione (7b) 138

3-(4-Chlorophenyl)-1H-isochromene-1-thione (7c) 139

3-(2-Bromophenyl)-1H-isochromene-1-thione (7d) 139

3-(3-Iodophenyl)-1H-isochromene-1-thione (7e) 139

3-(2,4-Dichlorophenyl)-1H-isochromene-1-thione (7f) 140

3-(2-Chloro-4-fluorophenyl)-1H-isochromene-1-thione (7g) 140

3-(4-Methoxyphenyl)-1H-isochromene-1-thione (7h) 140

3-(4-Fluorobenzyl)-1H-isochromene-1-thione (7i) 140

3-(Pentadecyl)-1H-isochromene-1-thione (7j) 141

7.5 Results and Discussion 141

xii

7.6 Biological Activities 144

7.7 Antibacterial Activity 144

Synthesis of (±)-1-aryl-7,8-dichloro-3,4-dihydro-

1H-isochromenes 150

7.8 Introduction 150

7.9 Experimental 151

General procedure for the synthesis of (±)-1-aryl-7,8-

dichloro-3,4-dihydro-1H-isochromenes (2a-g) 151

7,8-Dichloro-1-phenyl-3,4-dihydro-1H-isochromene (2a) 151

7,8-Dichloro-1-(2-chlorophenyl)-3,4-dihydro-1H-isochromene(2b) 152

7,8-Dichloro-1-(4-chlorophenyl)-3,4-dihydro-1H-isochromene (2c) 152

7,8-Dichloro-1-(3-methoxyphenyl)-3,4-dihydro-1H-isochromene(2d)152

7,8-Dichloro-1-(3-methoxy-4-hydroxyphenyl)-3,4-dihydro-

1H-isochromene (2e) 153

7,8-Dichloro-1-(3,4,5-trimethoxyphenyl)-3,4-dihydro-

1H-isochromene (2f) 153

7,8-Dichloro-1-(5-nitrobenzo[d] [1,3]dioxol-6-yl)-3,4-dihydro-

1H-isochromene (2g) 153

7.10 Results and Discussion 154

7.11 Antibacterial Activity 156

References Part II 159

xiii

List of Tables

Table 1.1 Comparison of the melting points of isocoumarins

and dihydroisocoumarins 4

Table 3.1 Physical constants and FTIR spectral data of the compounds (1-12) 59

Table 3.2 1H and

13C NMR data of the compound (9) 60

Table 3.3 1H and

13C NMR data of the compound (10) 61

Table 3.4 1H and

13C NMR data of the compound (11) 63

Table 3.5 1H and

13C NMR data of the compound (12) 64

Table 3.6 Physical constants and FT-IR spectral data of the compound (13) 66

Table 3.7 1H and

13C NMR data of the compound (13) 66

Table 3.8 Physical constants and FTIR spectral data of the compounds (16a-j)68

Table 3.9 Elemental analysis data of the compounds (16a-j) 68

Table 3.10 1H and

13C NMR data of compound (16a) 69

Table 3.11 1H and

13C NMR data of compound (16e) 70

Table 3.12 1H and

13C NMR data of compound (16h) 71

Table 3.13 Physical constants and FTIR spectral data of the

compounds (17a-j) 74

Table 3.14 Elemental analysis data of the compounds (17a-j) 74

Table 3.15 1H and

13C NMR data of compound (17a) 75

Table 3.16 1H and

13C NMR data of compound (17f) 76

Table 3.17 Physical constants and FTIR spectral data of the compounds (18a-j)78

Table 3.18 Elemental analysis data of the compounds (18a-j) 78

Table 3.19 1H and

13C NMR data of compound (18a) 79

Table 3.20 1H and

13C NMR data of compound (18e) 80

Table 3.21 1H and

13C NMR data of compound (18f) 81

Table 3.22 Physical constants and FTIR spectral data of the

compounds (19a-j) 83

Table 3.23 Elemental analysis data of the compounds (19a-j) 84

Table 3.24 1H and

13C NMR data of compound (19a) 84

Table 3.25 1H and

13C NMR data of compound (19e) 86

Table 3.26 1H and

13C NMR data of compound (19f) 87

xiv

Table 4.1 In vitro Antibacterial Activity of Isocoumarins (16a-j) 90

Table 4.2 In vitro Antibacterial Activity of Keto acids (17a-j) 91

Table 4.3 In vitro Antibacterial Activity of 3,4-Dihydroisocoumarins (18a-j) 92

Table 4.4 In vitro Antibacterial activity of 6,8-Dihydroxy3,4-

dihydroisocoumarins (19a-j) 93

Table 4.5 Antimalarial Activity of Isocoumarins (16a-j) 95

Table 4.6 Antimalarial Activity of Keto Acids (17a-j) 96

Table 4.7 Antimalarial Activity of 3,4-Dihydroisocoumarins (18a-j) 97

Table 4.8 Antimalarial Activity of 6,8-Dihydroxy-3,4-

Dihydroisocoumarins (19a-j) 98

Table 5.1 Physical and FTIR spectral data of the compounds (1-8) 113

Table 5.2 1H and

13C NMR data of Stellatin (8) 114

Table 5.3 In vitro antibacterial activity of compounds (5-8) 117

Table 7.1 Physical constants and FTIR spectral data of isocoumarins (4a-j) 135

Table 7.2 Physical constants and FTIR spectral data of isoquinolones (5a-j) 137

Table 7.3 Comparison of the δ values of H-4 among isocoumarins

and isoquinolones 137

Table 7.4 Comparison of the chemical shifts of H-4 and C-1

in compounds (6a-j) and (7a-j) 142

Table 7.5 Physical constants and FTIR spectral data of the compounds (2a-j) 143

Table 7.6 In vitro antibacterial activity of 3-substituted isocoumarins (4a-j) 146

Table 7.7 In vitro antibacterial activity of 3-substituted isoquinolones (5a-j) 146

Table 7.8 In vitro antibacterial activity of 3-substituted-1-

thioisocoumarins (7a-j) 147

Table 7.9 Physical constants and FTIR spectral data of the isochromans (2a-g)155

Table 7.10 In vitro antibacterial activity of isochromanes (2a-g) 157

xv

List of Figures

Fig. 3.1 Mass fragmentation pattern of the compound (9) 61

Fig. 3.2 Mass fragmentation pattern of the compound (10) 62

Fig. 3.3 Mass fragmentation pattern of the compound (11) 63

Fig. 3.4 Mass fragmentation pattern of the compound (12) 65

Fig. 3.5 Mass fragmentation pattern of the compound (16a) 70

Fig. 3.6 Mass fragmentation pattern of the compound (16e) 71

Fig. 3.7 Mass fragmentation pattern of the compound (16h) 72

Fig. 3.8 Mass fragmentation pattern of the compound (17a) 76

Fig. 3.9 Mass fragmentation pattern of the compound (17f) 77

Fig. 3.10 Mass fragmentation pattern of the compound (18a) 80

Fig. 3.11 Mass fragmentation pattern of the compound (18e) 81

Fig. 3.12 Mass fragmentation pattern of the compound (18f) 82

Fig. 3.13 Mass fragmentation pattern of the compound (19a) 85

Fig. 3.14 Mass fragmentation pattern of the compound (19e) 86

Fig. 3.15 Mass fragmentation pattern of the compound (19f) 87

Fig. 4.1 Cytotoxic activity results of the isocoumarins (16a-e) 101

Fig. 4.2 Cytotoxic activity results of the isocoumarins (16f-j) 101

Fig. 4.3 Cytotoxic activity results of the keto acids (17f-j) 102

Fig. 4.4 Cytotoxic activity results of the keto acids (17f-j) 102

Fig. 4.5 Cytotoxic activity results of the 3,4-dihydroisocoumarins (18a-e) 103

Fig. 4.6 Cytotoxic activity results of the 3,4-dihydroisocoumarins (18f-j) 103

Fig. 4.7 Cytotoxic activity results of the 6,8-dihydroxy-

3,4-Dihydroisocoumarins (19a-e) 104

Fig. 4.8 Cytotoxic activity results of the 6,8-dihydroxy-

3,4-Dihydroisocoumarins (19f-j) 105

Fig. 5.1 Mass fragmentation pattern of the Stellatin (8) 115

Fig. 5.2 Cytotoxic activity of the samples (5-8) 118

Fig. 7.1 Comparison of the antibacterial activity of 3-phenylsubstituted

isocoumarins (4a-j), 3-phenylsubstituted isoquinolin-

1(2H)-ones (5a-j) and 3-substituted phenyl-1H-isochromenes-

xvi

1-thiones (7a-j) 149

SCHEMES

Scheme 3.1 Synthesis of methyl 3,5-dimethoxy-4-methylphenyl acetate (9) 58

Scheme 3.2 Synthesis of 3,5-dimethoxy-4-methyl homophthalic acid (12) 59

Scheme 3.3 Synthesis of 6,8-dimethoxy-7-methyl-3-alkyl/arylisocoumarins (16a-j)67

Scheme 3.4 Synthesis of 6,8-dimethoxy-7-methyl-3-alkyl/aryl-

3,4-dihydroisocoumarins (18a-j) 73

Scheme 3.5 Synthesis of 6,8-dihydroxy-7-methyl-3-alkyl/aryl-

3,4-dihydroisocoumarins (19a-j) 83

Scheme 5.1 Synthesis of Stellatin 112

Scheme 7.1 Synthesis of 3-phenyl substituted isocoumarins (4a-j) 135

Scheme 7.2 Synthesis of 3-phenyl substituted isoquinolones (5a-j) 136

Scheme 7.3 Solvent-free conversion of isocoumarin into 1-thioisocoumarins 142

Scheme 7.4 Synthesis of isochromanes 155

1

INTRODUCTION

Isocoumarins and 3,4-dihydroisocoumarins are the secondary metabolites1 of a

wide variety of fungi, lichens, molds, bacteria, higher plants and insects. Majority of

isocoumarins have been isolated from various species of fungal genera Artemisia,

Aspergillus, Ceratocystis, Fusarium, Penicillum, Streptomyces, etc. A number of them

are constituents of a few higher plant families, e. g., Bignoniaceae, Compositae,

Leguminoseae, Myricaceae, Saxifragaceae. Literature reviews published on isocoumarins

include the review by R. D. Barry2 (1964), W. B. Turner and Aldridge

3 (1983), M.

Yamato4 (1983), R. A. Hill

5 (1986), E. Napolitano

6 (1997) and Bin

7 et al. (2000).

1.1 Nomenclature and Structural Type

The name isocoumarin (1) is derived from the fact that these compounds are

isomeric to coumarin (2). Coumarin8 was isolated (1820) from tonka tree formerly known

as Coumarouna odorata. In an isocoumarin, a lactonic pyran ring is fused to a benzene

ring. The IUPAC and Chemical Abstract name for isocoumarin is 1H-2-benzopyran-1-

one, numbered as shown and its 3,4-dihydro analogue (3) is named as 3,4-

dihydroisocoumarin rather than isochroman-1-one.

O

O

R2

R1

R3

R4

R5

R6

O O1

2

34

4a5

6

7

8 8aO

O

R2

R1

R3

R4

R5

R6

12

34

4a5

6

7

8 8a

(1) (2) (3)

As in case of other classes of the natural products (alkaloids, flavonoids, etc.) no

systematic nomenclature exists for isocoumarins. Majority of naturally occurring

isocoumarins and 3,4-dihydroisocoumarins have been assigned trivial names9 which are

derived from generic or specific names of source plant and fungi. Examples of the names

derived from those of parent genera are: agrimonolide (Agrimonia pilosa), fusamarin

(Fusarium spp.), alternariol (Alternaria spp.), artemidin (Artemisia glauca), peniolactol

(Peniophora sanguinea), cladosporin (Cladosporium spp.), homalicine (Homalium

zeylancum), oosponol (Oospora astringes), etc. Those names derived from species are

found in mellein (Aspergillus melleus), ustic acid (A. ustus), duclauxin (P. duclauxi),

2

ochratoxin A, B and C (A. ochraceus), capillarin (Artemisia capillaris), viridotoxin (A.

virinutans), moncerin (H. monoceros), etc.

Trivial names of a large number of isocoumarins end in the suffix "-in" for

example artemidin, bergenin, bactobolin A, B and C, actinobolin, baciphelacin,

coriandrin, asperentin, canescin, fusamarin, mellein, stellatin, etc. However, isocoumarin

names ending in other suffixes like “-ol, -one, -ide. -oic acid, anhydride” indicating their

chemical class are also common. Examples are altenuisol, hydrangenol, oosponol,

oospoglycol, peniolactol, reticulol, oospolactone, agrimonolide, feralolide, monocerolide,

ustic acid, β-callatolic acid, β-alectoronic acid, ardisic acid B, chebulic acid, lamellicolic

anhydride, naphthalic anhydride, etc.

Isocoumarin (1) itself (R1-R

6=H) has never been found to occur naturally,

however, its simple derivatives are found in nature. Isocoumarin may be substituted

either on lactone ring or the aromatic ring or on both. Thus R1-R

6 in (1) or (3) may be

alkyl, aryl, heterocyclyl, halo, nitro or any other substituent.

A number of naturally occurring isocoumarins possess a C-3 carbon substituent

and all isocoumarins, biogenetically derived from acetate have C-8 oxygenation and

some have retained the C-6 oxygen. Hydrangenol, phyllodulcin, chebulic acid,

dihydrohomalicine and blepherigenin are isocoumarins found in plants, lack C-6

oxygenation and are not acetate derived. Isocoumarins having a C-4, C-5 or C-7

substituents are relatively uncommon in nature, nevertheless, C-7 oxygenation is fairly

uncommon.

Mellein (4), the 3,4-dihydro-8-hydroxy-3-methylisocoumarin has been taken as

the parent compound for simple isocoumarins. Thus, 3,4-dihydro-8-hydroxy-6-methoxy-

3-methylisocoumarin (5) is known as 6-methoxymellein. Similarly, the compounds (6,

R1=H, R

2=COOH) and (6, R

1=CHO, R

2=H) are called as 7-carboxymellein and 5-

formylmellein, respectively.

O

O

CH3

OH

(4)

O

O

CH3

OH

H3CO

(5)

O

O

CH3

OH

R2

R1

(6)

3

Peniolactol (7a) and 3-alkyl-3-hydroxy-3,4-dihydroisocoumarins such as ustic

acid (8a) and its derivatives exist in tautomeric equilibrium between their keto acid forms

(7a & 8a) and lactol forms (7b & 8b) respectively.

O

O

C15H31

OH

OH

OH

(7a)

O

O

C15H31

OH

OHOH

(7b)

O

O

CH3

OH

OH

OH

OHOCH3

(8a)

O

O

CH3

OH

OHOH

OHOCH3

(8b)

The lactam analogue of isocoumarin, 1-(2H)-isoquinolinone (9a), trivially known

as isocarbostyril, exists in equilibrium with its tautomeric form (9b). A large number of

variously substituted isocarbostyrils10

and tetrahydroisoquinolinones (10) can also exist

as their other tautomers have been prepared.

NH

O

(9a)

RN

OH

(9b)

RNH

O

(10)

R

Sulphur analogues have also been known since long and a number of substituted

1-thio- (11, Z=S), 1-hydrazino-(11, Z=NNH2), 1-phenylhydrazino- (11, Z=NNHC6H5), 2-

thio- (12), and 1,2-dithioisocoumarins11

(13) have been prepared.

O

Z

OR

(11)

S

O

(12)

S

S

(13) In 1980, a three-step synthesis of 2-seleno- and 2-telluroisocoumarins was

reported12

. Regiospesific nucleophilic β-addition of methaneselenolate or -tellurolate

4

anion to the triple bond of ethyl-2-ethenylbenzoate (14) afforded the chalcogenated esters

(14a). Saponification afforded the corresponding acids (14b) which were electrophilically

cyclized via the acid chlorides to 1H-2-seleno-(15) and 1H-2-telluro- (16)-3-benzopyran-

1-ones.

O

OR X

O

14a) R= C2H5, Y= Se or Te

14b) R= H, Y= Se or Te

15) X= Se

16) X= Te

Y

1.2 Physical Properties

Isocoumarins are usually crystalline solids having higher melting points as

compared to corresponding 3,4-dihydroisocoumarins. Some of the isocoumarins like 3-

propylisocoumarin, 3,5-dimethyl-6,7-dimethoxy-8-hydroxyisocoumarin, etc. are oils.

Isocoumarins have melting points ranging from 49-50 °C (trans-artemidin) to 350 °C

(alternariol) but most of them melt point in the range: 120-180 °C. A comparison of the

melting points of isocoumarins and 3,4-dihydroisocoumarins is presented in Table 1.1.

Table 1.1 Comparison of the melting points of isocoumarins and

dihydroisocoumarins

Substitution

Melting points (°C)

Isocoumarin Dihydroisoumarin

6-methoxy 98 68

8-hydroxy 123-124 56-57

6,7-dimethoxy 122-123 140-141

6,8-dimethoxy-3-methyl 154-156 125-128

6,8-dimethoxy-3-pentadecyl 101 51-53

1.3 Biosynthesis:

The biosynthesis of simple dihydroisocoumarins such as mellein (4) and

derivatives has been firmly established by studies with 13

C-labeled acetate13

and 1H,

13C

doubly labeled acetate14

.

5

O

OH O

CH3

(4)

A pentaketide chain is formed by the condensation of five acetate units which

then undergoes reduction of the end keto group to an alcohol (17) followed by cyclization

allowing lactone formation in a diphenolic dihydroisocoumarin system (18). Similar

pentaketide chain after selective reduction of the central keto group as well as the end

group gives (19) which through β-elimination of hydroxyl group results in the formation

of deoxypentaketide (20). Cyclization can give a monophenol lactonemellein (4)

(Scheme 1.1).

O

OH O

CH3

CH3COONaOH

OO

O CH3

O O

O

OH O

CH3OHOH

OO

O CH3

O OH

Pentaketide (17) (18)

OH

OO

OH CH3

O OH

(19)

OH

OO

CH3

O OH

(20) (4)

Scheme 1.1

Potent antiulcer antibiotics xenocoumacin I and xenocoumacin II, can be

considered to arise from leucine and four acetate units15

. They condense together to form

the polyketide (21) which through selective reduction and cyclization would give the

aminodihydroisocoumarin (22). Alternatively, cyclization of the polyketide (21) would

give the acylated orselinic acid (23) and then the isocoumarine (24). At some point,

deoxygenation at C6 and reduction at C3, C4 produces (22) (Scheme 1.2).

6

OH

OO

O

O O

NH2

CH3

CH3

(21)

OH

OH O

OH

CH3

CH3

NH2

O

(23)

O

OH O

CH3

CH3

NH2

OH

(24)

O

OH O

CH3

CH3NH2

(22)

Scheme 1.2

But xenocoumacin I is produced after acylation of the amino group of (22) by a

unit derived from acetic acid and an amino acid arginine. On the other hand when acetic

acid and praline acylate the amino group of (22), it produces xenocoumacine II. In both

the cases, oxidation occurs at C8 and reduction at C9 to produce the diol system (25)

(Scheme 1.3).

O

OH O

NH

CH3CH3

O O

NH2

R

[H]

[O]O

OH O

NH

CH3CH3

O OH

NH2

ROH

(25) Scheme 1.3

The isocoumarin halorosellins A (28) and B (29) might be biosynthesized from

pentaketide (26) via the intermediate (27) a naturally occurring isocoumarin, Sclerin A16

.

Dehydration followed by glycosylation of (27) gives rise to halorosellins A (28) and B

(29) (Scheme 1.4). Both these compounds inhibit the growth of malarial parasite

Plasmodium falciparum17

.

7

O

O

O O

CH2R'O

CH3

RO

RORO

OR

R'=R=H= (28)

R'=CH3, R=H= (29)

OH

OO

O CH3

O O

O

OH O

CH3OH

CH3

OH

(26) (27) (28 and 29)O

O

O O

CH2R'O

CH3

RO

RORO

OR

Scheme 1.4

Cytogenin (30), a well known antitumor antibiotic, was biosynthesized by the

incorporation of 14

C-acetate and 14

C-methionine precursors18

.

O

OH O

OCH3 OH

(30) 13

C-NMR studies indicate that five acetate units by head to tail condensation give

pentaketide as an intermediate which cyclizes to lactone (31). The methyl group at 6-

OCH3 of cytogenin is derived from methyl group of methionine. Two pathways are

suggested (A), in which methylation with methionine occurs prior to hydroxylation of

(33). (B), in which methylation with methionine takes place after hydroxylation of (32).

Experimental results indicate that pathway (A) is correct and this mode of formation of

isocoumarin skeleton and its methylation are similar to that of reticulol19

(Scheme 1.5).

8

CH3

OOCH3

O O

O

O

OOH

OH CH3

(31)

pentaketide

5 X CH3COONa

O

OOH

OHOH

O

OOH

H3CO CH3

O

OOH

H3COOH

methylation

with methionine Hydroxylation

methylation

with methionineHydroxylation

A B

(32) (33)

(30) Scheme 1.5

Biosynthesis of canescin (34) a metabolite of Aspergillus malignus involves an

isocoumarin (35) as an enzyme free intermediate produced by the polyketide synthase.

O

O

CH3

OH

OH

O

O

OCH3 (34)

9

Oxidation of one of the methyl group of (35) gives the aldehyde (36) which is the

later intermediate of this biosynthetic pathway. The aldehyde (36) then undergoes an

aldol condensation with oxaloacetate to give (37). Two pathways A and B were

suggested in the conversion of (37) to (40) by decarboxylation and then lactonization.

A degree of uncertainty is created by the existence of a possible cross linking

between potential intermediate (38) and (39). The alcohol (41) produced as a result of

reduction of keto group of (40). The alcohol then undergoes O-methylatation to give the

canescin (34) (Scheme 1.6). The alcohol (41) has been isolated from the culture medium

which suggests that methylation is a late step in this biosynthetic pathway20

.

O

O

CH3

OH

OH

O

O

OCH3(34)

CH3

OOSEnz

CH3

O O

O

O

OOH

OH CH3

CH3O

OOH

OH CH3

O

H

O

OOH

OH CH3

OH

O

O

OH

O

OH

O

OOH

OH CH3

O

O

OH

O

OOH

OH CH3

OH

O

O

OH

A

B

O

O

CH3

OH

OH

O

O

O(40) (39)

(37)(38)

(36)(35)

O

O

CH3

OH

OH

O

O

OH (41)

polyketide synthase

Scheme 1.6

10

The mycotoxin ochratoxin A and B (43) are produced by Aspergillus ochraceus.

Incorporation studies21-22

with singly and doubly labeled 13

C-acetate have confirmed that

the dihydroisocoumarin moiety of the important mycotoxin ochratoxin A (42) and B (43)

has a regular polyketide origin. Five acetate units condensed to give pentaketide

ochratoxin α (44) and ochratoxin β (45) with a one carbon addition at C-7 from

methionine.

The biotransformation of pentaketide intermediate ochratoxin β (45) in to both

ochratoxin A (42) and B (43) was efficient. But already chlorinated ochratoxin α (44) was

only biotransformed significantly (4.85%) in to ochratoxin A (42), indicating that

chlorination is only the penultimate biosynthetic step in ochratoxin A biosynthesis. This

was supported by poor (1.5%) conversion of radiolabelled ochratoxin B (43) in to

ochratoxin A (42). Experimental results showed that some ochratoxin B (43) may arise

by dechlorination of ochratoxin A23

.

O

OOHO

NH

CH3

Cl

OOH(42)

O

OOHO

NH

CH3

OOH (43)

5 X MeCOONa

methylation

O

OOHO

OH

CH3

Cl

(44)methylation

O

OOHO

OH

CH3

(45)

Scheme 1.7

11

1.4 Pharmacological Applications

Isocoumarins and 3,4-Dihydroisocoumarins are the secondary metabolites of

fungi, bacteria, plants and are insect venoms and pheromones. A huge number of them

have been isolated from fungi, lichens and bacteria. Some higher plants, insect and

marine organisms are also the rich source of these secondary metabolites. They exhibit a

broad range of pharmacological activities including antiallergic, antimicrobial,

immunomodulatory, antifungal, antiinflammatory, cytotoxic, and antiangiogenic24-30

.

The insecticides which selectively act on the insect GABA receptor are toxic to

insect but not mammals31

. A new 3,4-dihydroisocoumarin derivative (44) isolated from a

fungal culture extract (which was screened for its ability to inhibit the specific binding of

the noncompetitive antagonist [3H]EBOB to housefly head membrane) of Neosartorya

quadricincta. Compound (44) at 2.2μM inhibited [3H]EBOB binding by 65%. This novel

GABA receptor ligand might prove to be lead compound for the identification of new

insecticides acting at the insect GABA receptor32

.

O

OOH

OOH

O

O

OH(44)

Two new isocoumarin derivatives, stoloniferol A (45) and B (46) were isolated

from the ethyl acetate extract of the sea squirt-derived fungus, penicillium stoloniferum

and a halophilic fungus, penicillium notatum. Their cytotoxicities were evaluated against

the P388, BEL-7402, A-549 and HL-60 cell lines using the MTT method33

.

(46)

O

OOH

OH

(45)

O

OOH

OHOMe

A novel isocoumarin (47) was obtained from a marine fungus Alternaria tenuis

possesses an unusual 7-membered ring in the side chain. This compound exhibited

cytotoxicity against human malignant A375-S2 and human cervicial cancer Hela cells34

.

12

It shows structural similarities to amicoumacins and the xenocoumacins which displayed

antibacterial, antitumor and potent antiulcer activities35-37

.

O

OH O

NH

O OH

OH

NH

O

OH

O(47)

An isocoumarin antibiotic (48) was isolated from the culture broth of Bacillus sp.

The strain was isolated from a soil sample collected at Iriomoto Island. Compound (48) is

a basic substituted isocoumarin, active against Gram-positive and Gram-negative

bacteria. It also showed a strong cytotoxic activity against the lymphoid leukemia cell

lines L1210 and P388. The antitumor activity was determined in mice against P388 cells.

It exhibited weak antitumor activity in vivo38

.

O

OOH

NH

O

OH

OH

NH2

(48)

Endophytic fungi, now recognized as potential producers of novel secondary

metabolites, can be used as possible biocontrol agents and drugs39

.

Two new

isocoumarins avicennin A (49) and B (50) with two derivatives (51) and (52) were

isolated from the mangrove endophytic fungus from the South China Sea40

.

O

O

Cl

OH

O

OH

(49)

O

O

O

OH

O

(50)

O

O

O

OH

OH

(51)

O

O

O

OH

(52)

13

7-Hydroxyartemidin (53) isolated from the ethanol/water (50:50, V/V) extract of

Artemisia drucunculus L. leaves. This table vegetable has long been used in folk

medicine as a natural food cure for cleaning and diluting of blood and treatment of

dizziness and headache41

.

O

O

OH

(53)

Dihydroisocoumarin (54) isolated from aerial parts of a small shrubs Xyris

pterygoblephara showed aromatase inhibitory activity42

. Aromatase enzyme is a well-

established target for the chemoprevention of breast cancer.

O

O

O

O

O

(54)

A number of natural products with pharmacological activity required structural

modification to reduce their toxicity. Three natural isocoumarins paepalantine (55),

paepalantine 9-O-β-D-glucopyranoside (56), paepalantine9-O-β-D-allopyranosyl(1→6)

glucopyranoside (57) and two semisynthetic paeoalantine 9,10 acylated (58) and

paepalantine 9-OH-10-Methylated derivative (59) obtained from the capitula of

paepalantus bromelioides. The compound (55) has antimicrobial activity as well as

significant in vitro cytotoxic effects in the McCoy cell line. It was observed that the side

effects are reduced by substitution of the 9 and/or 10-OH group43

.

O

O

O

OR1 OR2

O

(55) = R1 = R2 = H (56) = R1 = glc, R2 = H

(57) = R1 = -glc 6 all, R2 = H (58) = R1 = R2 = Ac

(59) = R1 = H, R2 = Me

14

Five isocoumarin derivatives, paraphaeosphaerins A-C (60-62) and

chaetochiversin A (63) and chaetochiversin B (64) have been isolated from solid agar

cultures of two fungal strains Paraphaeosphaeria quadriseptata and Chaetomium

chiversii living in association with the Sonoran desert plants, Opuntia leptocaulis, and

Ephedra fasciculate. These compounds are biogenetically related to monocillin I and

radicicol44

.

(60) (61)

O

O

OH

OH

OH

HOH H

O

O

OH

OHO

HHOHH

(63)

(64)

O

O

OH

OH

OH

HOH H

Cl

(62)

O

O

OH

OH

H OHO

H HO

O

OH

OH

Cl

O

HHOHH

Acrosin is a serine-dependent proteolytic enzyme which is responsible in the

dispersal of acrosomal matrix and also helps the sperm in the fertilization of oocytes45

. In

human it has also been studied that high acrosin activity of semen is associated with

improved fertility46-48

. Biotinylated isocoumarin suicide inhibitors were found to be

useful in the determination of activable proacrosin/acrosin levels in cryopreserved bull

semen49

.

An isocoumarin derivative Cytogenin (65) isolated from Streptoverticillium

eurocidicum is a well known antibiotic. It shows antitumor activity50

against Ehrlich

carcinoma at 6.3 to 100mg/Kg/day. It has also been demonstrated that cytogenin is

considerably effective as an immunological regulator51

.

15

O

O

OH

OH

O

(65)

Ascomycetes and basidiomycetes are the rich source of chlorine containing

secondary metabolites; most of them are pharmacologically active52

. Lachnum

papyraceum produce mycorrhizins and lachnumon type antibiotics. Production of these

antibiotics was strongly inhibited when the culture media of Lachnum papyraceum

treated with CaBr2 and six isocoumarins derivatives (66-71) having nematicidal and

antimicrobial activity were isolated53

.

O

OOH

R1O

R2 R3

R1 = R2 = R3 = H = (66) R1 = R2 = H, R3 = Cl = (67)

R1 = R2 = H, R3 = Br = (68) R1 = CH3, R2 = R3 = H = (69)

R1 = CH3, R2 = H, R3 = Cl = (70) R1 = H, R2 = OH, R 3 = Cl = (71)

Three novel dihydroisocoumarin derivatives (72-74) with alkyl substitution at

position 7 have been isolated from an endophytic fungus, Geotrichum sp., collected from

Crassocephalum crepidioides. All of these compounds possess antimalarial, antifungal

and antituberculous activity54

.

O

OOH

OH

(72)

O

OOH

OH

(73)

O

OOH

OH

(74)

2-(8-hydroxy-6-methoxy-1-oxo-1H-2-benzopyran-3-yl)propionic acid (NM-3)

(75) is a novel synthetic analogue of cytogenin, an isocoumarin isolated from culture

16

filtrate of Streptoverticillium eurocidium55-56

. NM-3 potently inhibits endothelial cell

proliferation, migration, sprouting, tube formation in vitro, and tumor growth in vivo57

. A

phase I clinical study of NM-3 in patients with cancer has demonstrated that it is a highly

orally bioavailable and well tolerated drug in humans58

.

O

OOH

O O

OH

(75)

One of the mechanisms involved in the progression of diabetic nephropathy, the

most common cause of end stage renal failure, is angiogenic phenomenon associated with

the increase of angiogenic factors such as vascular endothelial growth factor (VEGF)-A

and angiopoietin (Ang)-2, an antagonist of Ang-1. NM-3 significantly suppressed the

increase of VEGF induced by high glucose in cultured podocytes and also suppressed the

increase of VEGF and TGF-β induced by high glucose in cultured mesangial cells. This

reflects the potential use of NM-3 as a novel therapeutic agent for renal alterations in type

2 diabetes59

.

NM-3 also induces lethality of human carcinoma cells by both apoptotic and

nonapoptotic mechanism and potentiates the effects of cytotoxic chemotherapeutic

agents. NM-3 potentiates dexamethasone-induced killing of both dexamethasone-

sensitive multiple myeloma (MM1.S) and dexamethasone-resistant RPMI8226 and U266

multiple myeloma cells60

.

Urokinase-type plasminogen activator (uPA) is an attractive target for the

development of new compounds for its inhibition because uPA plays a major role in

extracellular proteolytic events associated with tumor cell growth, migration and

angiogenesis. uPA catalyzed the hydrolysis of extracellular plasminogen to plasmin. The

increased production of plasmin leads to the degradation of extracellular matrix, thereby

assisting the directional migration of cancer cells61-62

. uPA in complex with its receptor

uPAR also affects other boplogical processes including signaling pathways that

influence cell proliferation63

. Potent uncharged inhibitors of uPA can be developed based

upon isocoumarin scaffold. Bromine in the three position and an aromatic group in the

17

seven position are important contributors to binding. N-[3-(3-bromopropoxy)-4-chloro-1-

oxo-1H-isochromen-7-yl]benzamide (76) was identified as an uncharged lead inhibitor of

uPA64

.

O

O

O Br

Cl

NHO

(76)

1.5 Synthesis of Isocoumarins and 3,4-Dihydroisocoumarins:

A wide variety of synthetic approaches have been employed towards the synthesis

of isocoumarins65

or their 3,4-dihydro derivatives and a number of new methods are

being developed and reported each year66-74

. Isocoumarins and their 3,4-dihydro

analogues can be interconverted into each other. Following are some of the most

important highyield reactions used for the synthesis of isocoumarins and 3,4-

dihydroisocoumarins:

1.5.1 Oxidation of Isochromans:

Isochromans are oxidized into 3,4-dihydroisocoumarins in the presence of

selenium oxide, chromium oxide, potassium permanganate, nitric acid or air (Scheme

1.8).

O

R

O

O

R

(Scheme 1.8)

Isochroman prepared by 2-arylethanol75

upon oxidation in the presence of

pyridiniumchlorochromate and boiling dichloromethane76

gives 3,4-dihydroisocoumarins

which then can be converted into isocoumarins (77) (R=H, 7-CH3, 5-CF3, 5,6-C4H4) by

treating with n-bromosuccinimide and triethyl amine (Scheme 1.9).

18

R'O

(R' = H, MEM)

RO

R

O

O

RO

O

R

a) TiCl4 b) PCC c) NBS, (C 2H5)3N

(77)

(Scheme 1.9)

1.5.2 Oxidation of Indenes, Indanones and Indones:

It is the one of the most convenient, high yield general route for the synthesis of

isocoumarins and 3,4-dihydroisocoumarins. Indene (78) has been converted into

isocoumarins and 3,4-dihydroisocoumarins by their ozonization in ethanol, followed by

decomposition of the intermediate cyclic perester77

(79). The 2-carboxyphenyl-

acetaldehyde (80) lead to the formation of isocoumarins when treated with mineral acid

or copper powder78

and 3,4-dihydroisocoumarins when subjected to sodium borohydride

reduction (Sheme 1.10).

O

O

OH

OCH3

O

O

O

OCH3

CH3Alk

O

O

OH

O

O

O

O

C2H

5OH

NaBH4

O3

H+

(78)(79)

(80)

(Scheme 1.10)

6,8-Dioxygenated-3-alkyl substituted isocoumarins have been synthesized by

oxidative cleavage of indanone79-80

. First the 2-methylindan-1-one (81) was converted in

to silyl ether (82) which produced 2-hydroxyindanone (83) by ozonolysis. The 2-

hydroxyindanone on periodate cleavage afforded the isocoumarin (85) via the keto acid

(84). The 2-methylindan-1-one (81) can also be converted in to desired isocoumarin (85)

through enol (84a) followed by its ozonolysis (Scheme 1.11).

19

OO

CH3

R

CH3

(81)

OSi(CH 3)3OCH3

R

CH3

(82)

OSi(CH 3)3OCH3

R

CH3

O

OCH3

R

CH3

OH

O

(85)

OCOCF 3OCH3

R

CH3

(48a)

OCH3

R

O

CH3

O

OH

(84)OCH3

R

O

CH3

O

(83)

(Scheme 1.11)

Indanone have been converted in to indanone epoxide (86) by epoxidation with

H2O2/(C2H5)3N in acetone. The resulting epoxide was then submitted to flash vacuum

pyrolysis81

(FVP) (450 °C/0.1 mm) which undergoes rearrangement during FVP afforded

isocoumarins82

(87) (Scheme1.12).

O

O

CH3

OCH3

(C2H

5)N, H

2O

2

O

O

CH3

OCH3

O

FVP

(86)

O

CH3

O

OOCH3

O

OO

CH3

CH3

O

(87)

(Scheme 1.12)

20

1.5.3 Synthesis involving metals:

A number of methods have been reported in literature which involve synthesis of

isocoumarins and 3,4-dihydroisocoumarins by metallation (Lithiation, Silylation and

Thallation) at specific positions.

a) Lithiation:

This method was first discovered by Hauser83

and then extensively studied by

Narasimhan and Bhide84

. Benzoic acid derivatives are important precursors of

isocoumarins. Among the methods available for introducing a β-functionalized carbon

substituent ortho to the carboxyl group, those involving ortho-metallation of the benzene

ring have enjoyed a great popularity.

This approach has been thoroughly reviewed85-87

. Summarizing the general

concepts, carboxylic acid derivatives suitable for promoting ortho lithiation88-89

are

tertiary amides (4,4-dimethyl)oxazolin-2-yl group and secondary amides. Lithiated

tertiary amides are readily and generally ortho-lithiated using n-butyllithium and

tetramethylethylenediamine, but their reaction with alkylating agents other than methyl

iodide gives low yields because of a poor nucleophilicity.

Allylation of lithiated tertiary benzamides has, however, been accomplished in

high yields by previous trans-metallation to a magnesium or (better) to a copper

derivative; the allyl group thus introduced has been converted to the β-hydroxyalkyl

group required to complete the lactone ring in the conditions of the acid hydrolysis of the

benzamide, leading to racemic 3,4-dihydroisocoumarins directly, apparently without the

possibility of isolating the intermediate allylbenzoic acids.

Alternatively, asymmetric hydroxylation of the double bond followed by

treatment with acids has been used to obtain 3,4-dihydroisocoumarins with a high degree

of enantiomeric purity, as demonstrated by the enantioselective synthesis of the

isocoumarin portion of AI77B (88) (Scheme 1.13)90-93

.

21

O

NCH3

CH3

OCH3 O

NCH3

CH3

OCH3

CH3

CH3

O

O

OCH3

CH3

CH3OH

O

O

OH

CH3

CH3NH2

(88)

a) BuLi, TMEDA b) CuCN (LiCl2) c) (E)-1-bromo-5-methyl-2-hexene

d) Sharples AD e) aq. NaOH and then HCl

a, b, c d, e

(Scheme 1.13)

Enantiomerically pure natural 3,4-dihydroisocoumarins have been obtained from

lithiated secondary benzamides and homochiral epoxides. Coupling between lithiated

secondary benzamides and epoxides belongs to the beginning of the anionic chemistry of

aromatic compounds; unfortunately, yields are generally modest and N-alkylation can

complicate the reaction94

. Good yields have occasionally been reported though, as in the

synthesis of the allergenic principle of gingko biloba (89) (Scheme 1.14)95

and of a

variety of mellein derivatives96

.

O

NHCH3

Li

OCH3 O

NHCH3

OCH3

OHH25C12

a b

O

O

OCH3

C12H25

c

O

O

OCH3

C12H25

(89)

70%

98%

a) (R)-1,2-epoxytetradecane

c) BBr3

b) -OH, then neutrallization CuCN (LiCl2)

(Scheme 1.14)

22

This method can be used for the synthesis of 5-methoxy, 6-methoxy, 8-methoxy,

3-methyl-8-methoxy, 3-methyl-8-hydroxy-3,4-dihydroisocoumarins. Mellein, Kigelin,

Hydrangenol, Phyllodulcin, 3-methyl, 6-methyl, 3,6-dimethyl, 6-chloro, 8-chloro etc

have also been been prepared by this method.

b) Thallation-olefination of Arenes:

Isocoumarins and 3,4-dihydroisocoumarins were prepared in a single pot

reaction97

by reacting a benzoic acid with an electrophilic thallium salt in the presence of

an organic solvent to give O-thalliated benzoic acid followed by reaction with an organic

compound e.g. an alkene in the presence of PdCl2 (Scheme 1.15).

O

OH

TI

CH2=CH2

PdCl2

O

OR R

(Scheme 1.15)

c) Silylation:

Closely related to lithiation is the desilylation of 2-(trimethylsilylmethyl)-

benzamides, which generates carbanions suitable for additions to aldehydes98

. 2-

(Trimethylsilylmethyl)benzoyl chloride (90) also undergo desilylation and addition to

aldehydes to give dihydroisocoumarins (91) through a concerted mechanism involving

ortho-quinodimethanes rather than carbanions as reactive intermediates (Scheme 1.16)99

.

CH3

O

OH

OCH3

a

O

OH

OCH3

Si(CH3)3 b

O

Cl

OCH3

Si(CH3)3 c

O

O

OCH3

Ph

CH2

OOCH3

a) n-BuLi, (CH3)3SiCl b) SOCl2 c) CsF, ArCHO

(90)

(91)

(Scheme 1.16)

23

To this class of reactive intermediates belongs the products of UV irradiation of

ortho-toluyl cyanides which add to aliphatic and aromatic acyl cyanides to give 3-cyano-

3-phenyl-8-methoxy-3,4-dihydroisocoumarins which are converted to isocoumarins by

treatment with strong bases (Scheme 1.17)100

.

CH3

OOCH3

CH3

OOCH3

CN

a, b cCH2

OHOCH3

CN

OOCH3

O

Ph

CN

a) (CH3)3SiCN b) PCC c) hv, PhCOCN

(Scheme 1.17)

d) Organo-mercury catalyzed synthesis:

A facile synthesis of 3-substituted arylisocoumarins involve the reaction of ester

(92) (R=H, Br, Cl, I, Ac)101

with mercuric acetate to give isocoumarin mercurials which

undergo substitution reactions to afford the isocoumarins (93) (R1=H, CH3, Cl, Br; R=H,

Br, Cl, I, Ac) (Scheme 1.18).

R

CO2Me

Hg(OAc)2

O

O

RR

1

(92) (93)

(Scheme 1.18)

Sulphuric acid-catalyzed chloralhydrate condensation with different m-substituted

benzoic acids formed trichlorophthalides (94), from which Zn+AcOH reduction afforded

various dichloro derivatives (95). These derivatives on treatment with alkaline Hg(OAc)2

+ I2 furnished different substituted isocoumarins (96a-c) (Scheme 1.19)102

.

24

O

O

R

CCl 3

Zn-AcOH

COOHR

Cl

Cl

Hg(OAc)2

NaHCO3, I2

DMSO

70-80 °C, 3-4hR

Cl

O

O

96a), R= OCH3

96b), R= OH

96c), R= OCH2Ph

(94) (95)

(Scheme 1.19)

e) Palladium catalyzed method

Chemoselective reduction of the carboxylic acid (97) to the corresponding alcohol

and subsequent protection as the silyl ether afforded compound (98) in 87% overall yield.

A palladium catalyzed Heck-type coupling of orth Iodo benzoate with acylate derivative

provided the compound (99). Acid catalyzed intramolecular condensation of (99) in 5%

HCl/MeOH resulted in the formation of isocoumarin (100) (Scheme 1.20)103

.

I

OMe

MeO

O

OH

O

OMe b

I

OMe

MeO

TBSO

O

OMe c

OMe

MeO

TBSO

O

OMe

COOMeMeO

MeOO

O

OH

OMe

O

OMe

(97) (98)

(99)

(100)

b) i. BH3, THF, ii. TBSCl, imidazole,

c) Pd(PPh3)4, K2CO3 d) 5% HCl/MeOH

Scheme 1.20

f. Iridium catalyzed method

Two new cyclizations of ketoaldehydes104

have been developed using an Ir-ligand

bifunctional catalyst. Oxidative lactonization of δ-ketoaldehydes proceeded smoothly at

room temperature to give coumarin derivatives in excellent yields. Intramolecular

Tishchenko reaction of δ-ketoaldehydes afforded 3,4-dihydroisocoumarins (101a-b) in

good yields (Scheme 1.21).

25

O

R

O

R= CH3

R= Ph

O

R

O

OH

R

O+

O

R

O

O

R

O+

O

R

O

101a) R= CH3

101b) R= Ph

Ir Cat (5mol%)

t-BuOH, reflux

(5mol%), cooxidant

base, rt, 16h

NHIr

+

O

Ph

Ph

CH3

CH3CH3

CH3

CH3

Scheme 1.21

g. Rhodium-Catalyzed Oxidative Coupling of Benzoic Acids with Alkynes via

Regioselective C-H Bond Cleavage

The oxidative coupling of benzoic acids with internal alkynes effectively proceeds

in the presence of [Cp*RhCl2]2 and Cu (OAc)2 H2O as catalyst105

and oxidant

respectively to produce the corresponding isocoumarin derivatives. The copper salt can

be reduced to a catalytic quantity under air (Scheme 1.22).

O

OH

H

+ R RRh-Cat

Cu-salt O

R

R

O

Scheme 1.22

1.5.4 Aldol-type Condensation between Homophthalic Acids, Esters or

Anhydrides and Carbonyl Compounds

Isocoumarins and 3,4-dihydroisocoumarins are most commonly synthesized by

using this type of condensation. The most important methods of aldol type condensation

are discussed in four main groups.

26

a. Stobbe Condensation of Homophthalates with Aldehydes and Ketones

Stobbe condensation is used for synthesis of a number of 3,4-dihydro-

isocoumarins106-110

. Synthesis of (dl)-agrimonolide111

provides a good example of

application of Stobbe condensation. Thus, diethyl 3,4-dibenzyloxyhomophthalate (102)

on condensation with 4-methoxybenzaldehyde in presence of sodium hydride afforded

2,4-dibenzyloxy-6-[1-ethoxycarbonyl-4-(4'-methoxyphenyl)buten-1-yl]benzoic acid

(103a, R=COOEt).

Hydrolysis and decarboxylation gave 2,4-dibenzyloxy-6-[4-(4'-methoxyphenyl)

buten-1-yl]benzoic acid (103b, R=H) which on cyclization with bromine gave the 4-

bromo-3,4-dihydroisocoumarin (104). Reductive debromination and debenzylation was

simultaneously effected by adding triethyl amine to the catalytic reduction medium to

furnish the (dl)-agrimonolide (105) (Scheme 1.23).

COOC 2H5

COOC 2H5

BzO

OBz

MeO

O COOH

BzO

OBz

ROMe

NaOH

COOH

BzO

OBz

OMe

Br2 / CHCl

3

BzO

OBz

OMe

O

O

Br

H2 / Pd-C / (C

2H

5)N

BzO

OBz

OMe

O

O

Br

(102) (103a)

(103b)

(104)

(105)

R= COOC2H5

R= H

Scheme 1.23

b. Claisen Condensation of Homophthalates with Formates

Diethyl homophthalate (106) condenses with methyl formate in the presence of

sodium ethoxide imparting a 66% yield of isocoumarin-4-carboxylic acid (107).

27

Decarboxylation with phosphoric acid furnishes isocoumarin (108) (Scheme 1.24)112

.

COOC 2H5

COOC 2H5

(106)

O

O

COOH

(107)

O

O(108)

HCOOCH3

C2H

5ONa

H3PO

4

-CO2

Scheme 1.24

6,7-Dimethoxyisocoumarin and 5,7-dimethoxyisocoumarin were also prepared by

the above procedure. Ethyl 5,6,7-trimethoxyisocoumarin-4-carboxylate was prepared

from corresponding homophthalate and ethyl formate in the presence of potassium

ethoxide in good yield113

.

c. Claisen Condensations of Homophthalates with Oxalates

Metallic sodium in ether or better without a solvent effects ready condensation

between diethyl homophthalate (109) and diethyl oxalate giving a 67% yield of the

triester (110). This triester loses ethanol when heated yielding diethyl isocoumarin-3,4-

dicarboxylate (111). Under different hydrolysis conditions different products are formed.

Thus heating (111) at 68-72°C for 3hr. gives ethyl isocoumarin-3-(carboxylic

acid)-4-carboxylate (112) and prolonged heating yields isocoumarin-3-carboxylic acid

(113). Boiling hydrochloric acid or heating in a sealed tube at 180-190°C converts (111)

to isocoumarin-3-carboxylic acid in 84% yield114

. These results indicate that the ester at

position 3 in (111) is hydrolyzed first, but the acid at position 4 is more easily

decarboxylated (Scheme 1.25).

28

COOC 2H5

COOC 2H5

(109)

O

O

COOC 2H5

COOC 2H5

(111)

NaO

OC2H5O

H5C2O

+COCOOC 2H5

COOC 2H5

O OC2H5

(110)

O

O

COOH

COOC 2H5

(112)

O

O

COOH

(113)

Scheme 1.25

d. Condensation of malonyl heterocycles withdiphenylcarbonate

Reaction of diphenylcarbonate with enolized phenylmalonyl heterocyclic compounds as

(114 a-d) yields the condensed isocoumarins115

like (115 a-d) (Scheme 1.26).

X

OH

O

(114a-d)

O

OC6H5H5C6O

X

O

O

O

(115a-d)

a) X= H

b) X= NH

c) X= NCH3

d) X= NC6H

5

X is same as in 114a-d

Scheme 1.26

e. Condensation of Acid chlorides, Phenols, Phenol acids with homophthalic

acids and Anhydrides

Tirodkar and R. N. Usgaonkar116-117

carried out two or three step synthesis of

various 3-alkyl/aryl isocoumarins. The synthesis involved pyridine catalyzed acylation of

homophtalic acids with acid chlorides or anhydrides to give isochroman-1,3-dione (116).

29

Treatment of (116) with conc. sulphuric acid at room temperature gave the 3-alkyl/aryl

isocoumarin-3-carboxylic acid whereas on treatment with 90% sulphuric acid at 90°C

directly gave the isocoumarins (Scheme 1.27).

COOH

COOH

R

(R'CO)2O / Py

R'

O

O

COOH

R

r.t

O

O

O

COR'

R

R'

O

O

R

Conc. H2SO4

90% H2SO

4

90 °C

(116)

Scheme 1.27

S. Nakajima et. al. synthesized various 3-arylisocoumarins (118, Ar =Ph, p-

anisyl, p-(OH)phenyl etc.) and later on 3-alkylisocoumarins in high yields (80%) by

heating directly the homophthalic acids (117, R, R1, R

2=H, OH, OMe, Cl) with aryl or

acyl chlorides at 190°C. These isocoumarins were converted into corresponding 3,4-

dihydroisocoumarins (Scheme 1.28).

COOH

COOH

R

R1

R2

Ar/R

O

R

R1

R2

O

Ar / RCOCl

190 °C

(117) (118)

Scheme 1.28

A. Rose118

and later on H. Yoshikawa119

prepared a large number of 3-

(hydroxyphenyl)isocoumarins by condensation of various phenols with substituted

homophthalic acids in moderate yields in presence of polyphosphoric acid or the

30

anhydrous stannic chloride e.g. 7-methyl-3-(2′-hydroxy-4′-methylphenyl)isocoumarin

(119) was obtained from 7-methylhomophthalic acid (Scheme 1.29).

COOH

COOH

CH3O

O

CH3

CH3

OH

(119)

Scheme 1.29

3-(2’,4’-Dimethoxyphenyl)-, 3-(2’-methyl-4’-hydroxyphenyl)isocoumarins etc.

were prepared120

by condensation of homophthalic anhydride with appropriate phenols.

3-(4'-Methoxyphenyl)isocoumarin (120) was prepared by condensation of homophthalic

acid with anisole (Scheme 1.30).

COOH

COOH

O

O

OCH3

(120)

PPA

OCH3

Scheme 1.30

1.6 Reactions of Isocoumarins and 3,4-dihydroisocoumarins

1.6.1 Hydrolysis

Isocoumarins are lactones and undergo ring opening on alkaline hydrolysis to

give homophthaldehydic acids or ketones (121, R’=H or alkyl, aryl) or hydroxyl acid

(122, R’=alkyl, aryl, etc.). Similar treatment of 3,4-dihydroisocoumarins yields the

corresponding β-(2-carboxyphenyl)ethyl alcohol. In most of the cases isolation of the free

acids due to spontaneous recyclization to lactonic ring is not possible. In some cases e.g.

during the hydrolysis of cis-3-phenyl-4-hydroxy-3,4-dihydroisocoumarin (123), the

glycol produced (124) recyclizes to the more stable erythro-γ-lactone (125) under acid

treatment (Scheme 1.31)121-122

.

31

COOH

R'

OR

COOH

R'

OHR

(121) (122)

Ph

O

O

OHHH

(123)

COOH

Ph

OH

OHHH

Ph

O

O

H

HOH

(125)(124)

Scheme 1.31

1.6.2 Reaction with Ammonia and Amines

Ammonia and amines add to isocoumarins furnishing isocarbostyrils123

(126), a

reaction typical of esters (Scheme 1.32).

O

O

NR

O

NH3

or

RNH2

(126)

Scheme 1.31

For example isocoumarin and 3-carboxylic acid have been condensed with

tryptamine, and the product subsequently converted to yobyrine (127) and other

derivatives (Scheme 1.32)124

.

O

O

NR

ON

(127)

+N

CH3

NH2

N

O

NH

Scheme 1.32

32

It is reported that 3,4-dihydroisocoumarin with ammonia gives the corresponding

tetrahydroisoquinolinones e.g. heating agrimonolide with ammonia at 100°C gave the

isoquinolinone analogue (128) (Scheme 1.33).

O

O

OMe

OH

OH

(128)

NH3

100 °CNH

O

OMe

OH

OH

Scheme 1.33

1.6.3 Reaction with Phosphorus Pentasulfide

Isocoumarin can be converted to 1-thioisocoumarin (129) with phosphorus

pentasulfide and treatment of 1-thioisocoumarin with ammonium sulfide or aniline yields

isoquinolins (Scheme 1.34). Analogously, 3-phenylisocoumarin has been converted to 1-

thio-3-phenylisocoumarin, and treatment with aniline produced (130)125

.

O

O

(129)

N

P2S

5

O

S

(130)

N

O

Ph

Ph

Scheme 1.34

1.6.4 Nitration

The only report of the nitration of an isocoumarin is that of 3-phenyl-3,4-

dihydroisocoumarin (131), in which nitric acid in sulfuric acid gives 3-(4-nitrophenyl)-7-

nitro-3,4-dihydroisocoumarin (132) (Scheme 1.35)126

.

O

O

(131)

HNO3

O

O

NO2

(132)

Scheme 1.35

33

1.6.5 Reaction with Grignard Reagents

Addition of phenylmagnesium bromide127-132

to 3-phenylisocoumarin followed by

perchloric acid, anhydrous hydrochloric acid, ferric chloride or ferric bromide yields the

isobenzopyrilium salt (133) (Scheme 1.35).

O

O

Ph

C6H

5MgBr

O

OH

Ph

Ph

O

Ph

Ph

(133)

HY

+Y

-

Y = perchlorate, chloride, ferric chloride or ferric bromide

Scheme 1.35

1.6.6 Oxidation

Chromium trioxide oxidation of 3,4,6,7-tetraphenylisocoumarin (134) produce 2-

benzoyl-4,5-diphenylbenzoic acid (135) (Scheme 1.36).

O

O

Ph

Ph

Ph

Ph

(134)

COOH

O

Ph

Ph

Ph

CrO3

(135)

Scheme 1.36

1.6.7 Reduction

The double bond present between C-3 and C-4 of isocoumarin can be reduced

readily with hydrogen and palladium on charcoal or with other catalyst133-134

. Catalytic

reduction also has been used to remove the halogen from cis- and trans-3-phenyl-4-halo-

3,4-dihydroisocoumarin135-136

.

1.7 Interconverision of isocoumarins and 3,4-dihydroisocumarins

It has been observed that some methods directly afford the isocoumarins while

others produce dihydroisocoumarins. Their interconversion is carried out depending upon

whether the synthesis of isocoumarin is easier or that of its dihydro derivative.

34

1.7.1 Conversion of 3,4-Dihydroisocoumarins to Isocoumarins

There are two routs mainly used for conversion of 3,4-dihydroisocoumarins to

isocoumarins:

a. Alkaline Hydrolysis Followed by Oxidation and Recyclization

Alkaline hydrolysis of 3,4-dihydroisocoumarins137

yields the hydroxy acids which

could be oxidized to corresponding keto-acids. Since the hydroxy acids on standing

recyclize to parent dihydroisocoumarins, the oxidation should be carried out immediately.

The keto-acids are readily cyclized e.g. by heating with acetic anhydride to corresponding

isocoumarins (Scheme 1.37).

O

O

R

R COOHOH

RCrO

3

R COOHO

R

R

O

O

R

R

Scheme 1.37

b. Benzylic Bromination Followed by Dehydrobromination

Isocoumarins can be prepared from 3,4-dihydroisocoumarin138-139

via benzylic

bromination with N-bromosuccinimide (NBS), followed by dehydrohalogenation with

triethylamine (Scheme 1.38).

O

O

MeO

MeO

O

O

MeO

MeO

Br

NBS

UV O

O

MeO

MeO(C

2H

5)

3N

Scheme 1.38

1.7.2 Conversion of Isocoumarins to 3,4-Dihydroisocoumarins

Two different methods of reduction mainly used for conversion of isocoumarins

to 3,4-dihydroisocoumarins:

a. Alkaline Hydrolysis Followed by Reduction and Recyclization

Alkaline hydrolysis of isocoumarins with dilute aqueous alkali affords the keto-

acids, which upon reduction with sodium borohydride are converted into corresponding

35

hydroxy-acids. Cyclodehydration of the latter affords the dihydroisocoumarins (Scheme

1.39).

O

OOMe

Ar/R

COOHO

Ar/R

OMe

KOH

MeOH

NaBH4

COOHOH

Ar/R

OMe

O

OOMe

Ar/R

-H2O

Scheme 1.39

b. Catalytic Reduction

Hydrogenation140-141

in the presence of palladium charcoal or some other catalyst

has been used to reduce the 3,4-double bond of isocoumarins thereby converting them

directly into 3,4-dihydroisocoumarins.

36

2. EXPERIMENTAL

2.1 Purification of Solvents

The solvents were purified and dried according to the standard procedures before

use. The dried solvents were stored under molecular sieves (4 Å).

2.2 Instrumentation

Melting points were recorded using a digital Gallenkamp (SANYO) model MPD

BM 3.5 apparatus and are uncorrected. FTIR spectra were recorded using an FTS 3000

MX spectrophotometer, 1H NMR and

13C NMR spectra were determined as CDCl3

solutions at 300 MHz on a Bruker AM-300 spectrophotometer, mass spectra (EI, 70eV)

on a GCMS instrument, and elemental analyses with a LECO-183 CHNS analyzer

Aigilent Technologies USA. All the compounds were purified by thin layer

chromatography using silica gel HF-254 from Merck.

Synthesis of compounds

2.3 methyl 4-methylbenzoate (1)

A stirred solution of p-toluic acid (13.6g, 100mmol) in dry methanol (75ml) was

treated drop wise with conc. H2SO4 (5ml). The mixture was refluxed for 8-9hrs. The

reaction was monitored by TLC. After the completion of reaction, mixture was

concentrated to 55ml and extracted with ethyl acetate (3x50ml). The extract was washed

with saturated brine, dried and concentrated to afford methyl 4-methylbenzoate (1) as

thick oil (14.21g, 94.79%); Rf: 0.7 (petroleum ether and ethyl acetate, 4:1); m. p. 34°C;

IR (KBr): 3012 (C-H), 1742 (C=O), 1562 (C=C) cm-1

.

2.4 3, 5-Dibromo-4-methylbenzoate (2)

Aluminum chloride anhydrous (11.92g, 89mmol) was added portion wise to

stirred, cooled methyl 4-methyl benzoate (1) (5g, 33.30mmol). Bromine (3.56ml) was

then added to the stirred mixture for 45min at such a rate to keep the temperature at or

below 20°C. Stirring was continued at room temperature for 30min and at 80-85°C for

1h. The mixture was cooled to 30°C and treated with methanol (55.5ml) during 30min

and then stirred overnight. The crude product was collected by filtration, washed with

cooled (10°C) methanol and crystallized from methanol at 10°C to afford methyl 3,5-

dibromo-4-methylbenzoate (2) (5.99g, 58.38%) as colorless crystals, Rf: 0.6 (petroleum

37

ether and ethyl acetate, 4:1); m. p. 82-84 °C; IR (KBr): 3021 (C-H), 1719 (C=O), 1559

(C=C) cm-1

.

2.5 3, 5-Dimethoxy-4-methyl benzoic acid (3)

A pyridine solution of methyl 3,5-dibromo-4-methylbenzoate (2) (4.62g,

15.0mmol) was added to a solution of sodium methoxide (2.07g, 90.0mmol) in dry

methanol and freshly prepared anhydrous copper (I) chloride (0.212g, 1.25mmol). The

reaction mixture was refluxed under nitrogen for 15h, cooled to room temperature and

filtered. The filtered cake was washed with warm methanol. The solution was refluxed

for 1h, cooled to room temperature and diluted with saturated brine (30ml). The mixture

was extracted with ethyl acetate (30ml), the extract discarded and the aqueous phase

acidified with cold, concentrated HCl (10ml) and then extracted with ethyl acetate (3x

20ml). The extract was washed with saturated brine, dried and evaporated. Crystallization

of residue from aqueous methanol (1:2) yielded 3,5-dimethoxy-4-methyl benzoic acid (3)

(2.5g, 85%); Rf: 0.4 (petroleum ether and ethyl acetate, 4:1); m. p. 210-212 °C; IR (KBr):

3213 (O-H), 3029 (C-H), 1731 (C=O), 1569 (C=C) cm-1

.

2.6 Methyl 3,5-dimethoxy-4-methylbenzoate (4)

3,5-dimethoxy-4-methyl benzoic acid (3) (2.5g, 12.5mmol) was dissolved in dry

methanol (15ml) and then concentrated sulphuric acid (1-2ml) was added. The mixture

was refluxed for 13-14hrs. The reaction was monitored by TLC. After the completion of

reaction, mixture was concentrated and extracted with ethyl acetate (3x15ml). The extract

was washed with saturated brine, dried and evaporated to afford methyl 3, 5-dimethoxy-

4-methyl benzoate (4) (2.25g, 84%) as colorless crystals, Rf: 0.65 (petroleum ether and

ethyl acetate, 4:1); m. p. 76-78 °C; IR (KBr): 3019 (C-H), 1732 (C=O), 1571 (C=C) cm-1

.

2.7 (3, 5-Dimethoxy-4-methyl phenyl)methanol (5)

Methyl 3, 5-dimethoxy-4-methyl benzoate (4) (4.2g, 0.02 mol) and sodium

borohydride (4.5g, 0.12 mol) were suspended in freshly distilled THF (150ml). The

reaction mixture was stirred for 15min at 65°C and then added methanol (150 ml) drop

wise for 30min. The mixture was refluxed for 4hrs then cooled to room temperature and

treated with saturated ammonium chloride solution (150ml). Stirring was continued for

1hr then acidified with dilute hydrochloric acid and extracted with ethyl acetate

(3x20ml). The extract was dried, evaporated and (3,5-dimethoxy-4-methyl

38

phenyl)methanol (5) product was recryatallized with petroleum ether to afford prism like

crystals (3.0g, 82.41%); Rf: 0.5 (petroleum ether and ethyl acetate, 4:1); m. p. 45-47 °C,

IR (KBr): 3442 (O-H), 3023 (C-H), 1554 (C=C) cm-1

.

2.8 3, 5-Dimethoxy-4-methyl benzyl bromide (6)

3, 5-Dimethoxy-4-methyl benzyl alcohol (5) (10.01g, 55.0mmol) was dissolved in

dry benzene (40-50ml). The solution was treated with phosphorous tribromide (14.89g,

5.2ml, 55.0mmol) and stirred the resulting mixture for 4hrs. Then poured the reaction

mixture onto ice cold water, separated the organic layer and evaporated to afford crude 3,

5-Dimethoxy-4-methyl benzyl bromide (6). Prism like crystals were obtained after

recrystallization in petroleum ether (11.2g, 84.21%); Rf: 0.6 (petroleum ether and ethyl

acetate, 4:1); m. p. 68-69 °C, IR (KBr): 3013 (C-H), 1571 (C=C) cm-1

.

2.9 (3, 5-Dimethoxy-4-methylphenyl)acetonitrile (7)

3, 5-Dimethoxy-4-methyl benzyl bromide (6) (7.0g, 28.6mmol) was dissolved in

a mixture of ethyl alcohol (120ml) and water (120ml). Potassium cyanide (2.6g,

40.7mmol) was then added to reaction flask and refluxed for 4hrs. Reaction mixture was

poured onto ice cold water and extracted with ethyl acetate (3x20ml). The extract was

dried over anhydrous Na2SO4, evaporated to afford (3,5-dimethoxy-4-

methylphenyl)acetonitrile (7) and recrystallized in petroleum ether to get prism like

crystals (4.6g, 84.09%); Rf: 0.5 (petroleum ether and ethyl acetate, 4:1); m. p. 48-49 °C,

IR (KBr): 3007 (C-H), 1561 (C=C) cm-1

.

2.10 (3, 5-Dimethoxy-4-methylphenyl) acetic acid (8)

(3, 5-Dimethoxy-4-methylphenyl)acetonitrile (7) (4.0g, 20.9mmol) was dissolved

in a mixture of water (17.8ml) and dioxane (17.8ml). Then added potassium hydroxide

(14.56g in 15ml H2O) and refluxed the mixture for 10-12hrs. Reaction mixture was

poured onto ice cold water and extracted with ethyl acetate (20ml). The extract was

discarded and aqueous layer was acidified with dilute hydrochloric acid. Precipitates

were filtered to afford (3, 5-dimethoxy-4-methylphenyl) acetic acid (8) (3.1g, 70.61%);

Rf: 0.4 (petroleum ether and ethyl acetate, 4:1); m. p. 129-130 °C; IR (KBr): 3242 (O-H),

3013 (C-H), 1707 (C=O), 1567 (C=C) cm-1

.

39

2.11 Methyl (3, 5-dimethoxy-4-methyl phenyl) acetate (9)

A stirred solution of (3, 5-dimethoxy-4-methylphenyl) acetic acid (8) (5.0g,

23.8mmol) in dry methanol (30ml) was treated dropwise with conc. H2SO4 (5ml). The

mixture was refluxed for 8-9hrs. The reaction was monitored by TLC. After the

completion of reaction, mixture was concentrated to 55ml and extracted with ethyl

acetate (3x50ml). The extract was washed with saturated brine, dried and concentrated to

give crude oil which was distilled to afford methyl (3, 5-dimethoxy-4-methyl phenyl)

acetate (9) (4.7g, 88.18%), Rf: 0.7 (petroleum ether and ethyl acetate, 4:1); m. p. 38-40

°C; IR (KBr): 3023 (C-H), 1734 (C=O), 1573 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm):

7.45 (2H, s, H-2, H-6), 3.96 (6H, s, 2OCH3), 3.54 (2H, s, Ar-CH2), 3.47 (3H, s,

COOCH3), 2.55 (3H, s, Ar-CH3); 13

C NMR (CDCl3 δ ppm): 168.23 (C=O), 132.54 (C3,

C5), 128.37 (C2, C6), 119.42 (C4), 112.21 (C1), 68.55 (Ester OCH3), 55.34 (Ar-OCH3),

36.91 (CH2), 28.63 (Ar-CH3); MS (70eV): m/z (%); 224 [M]+.

(46), 193 (43), 165 (100),

59 (12); Anal. calcd for C12H16O4: C, 64.28 H, 7.14 O, 28.57 Found: C, 64.02 H, 6.96 O,

28.35.

2.12 Methyl (2-formyl-3, 5-dimethoxy-4-methyl phenyl) acetate (10)

Phosphorus oxychloride (1.61g, 10.0mmol) was added dropwise in to a stirred

solution of methyl (3, 5-dimethoxy-4-methyl phenyl) acetate (9) (2.0g, 8.9mmol) in dry

DMF (10ml) at 55 °C. Reaction mixture was heated at about 100 °C for 2hrs and stirred

overnight at room temperature. Then poured the reaction mixture into aqueous solution of

sodium acetate (10%, 10ml) and shake vigorously. Methyl (2-formyl-3, 5-dimethoxy-4-

methyl phenyl) acetate (10) was precipitated as yellowish solid (1.9g, 84%); Rf: 0.55

(petroleum ether and ethyl acetate, 4:1); m. p. 51-53 °C; IR (KBr): 3029 (C-H), 1722

(C=O), 1690 (CHO), 1545 (C=C) cm-1

; 1H NMR, (CDCl3, δ ppm ): 9.75 (1H, s CHO),

7.96 (1H, s, H-6), 3.42 (3H, s, 3-OCH3), 3.25 (3H, s, 5-OCH3), 3.11 (3H, s, CO2CH3),

2.92 (2H, s, Ar-CH2), 2.80 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm ): 179.32

(Aldehyde C=O), 162.43 (Ester C=O), 136.76 (C3, C5), 131.89 (C2), 126.21 (C6),121.33

(C4), 117.54 (C1), 61.63 (Ester OCH3), 57.34 (Ar-OCH3), 39.12 (Ar-CH2), 32.08 (Ar-

CH3); MS (70eV): m/z (%); 252 [M]+.

(25), 251 (65), 224 (49), 223 (34), 165 (100), 29

(31); Anal. calcd for C13H16O5: C, 61.90 H, 6.34 O, 31.74 Found: C, 61.67 H, 6.16 O,

31.56.

40

2.13 2,4-Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid (11)

Methyl (2-formyl-3, 5-dimethoxy-4-methyl phenyl) acetate (10) (6.3g, 25.0m

mol) and sulfamic acid (8.3g, 86.0mmol) in 150ml H2O:THF:DMSO (20:1:1) at 0°C was

treated with NaClO2 (7.24g, 80.0mmol) in 20ml H2O. The reaction mixture was stirred

for 20min at 0°C and then diluted with ethyl acetate (100ml), washed with saturated

aqueous ammonium chloride (2 x 130ml) and then with saturated aqueous sodium

chloride (130ml). Organic layer was dried over anhydrous sodium sulfate and evaporated

to afford 2,4-dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid (11) (6.6g,

79%); Rf: 0.4 (petroleum ether and ethyl acetate, 4:1); m. p. 164-166 °C; IR (KBr): 3265

(O-H), 3037 (C-H), 1734 (C=O), 1715 (COOH), 1562 (C=C) cm-1

; 1H NMR (CDCl3, δ

ppm ): 8.19 (1H, s, COOH), 7.66 (1H, s, H-6), 3.82 (3H, s, 3-OCH3), 3.67 (3H, s, 5-

OCH3), 3.63 (3H, s, CO2CH3), 2.54 (2H, s, Ar-CH2), 2.25 (3H, s, Ar-CH3); 13

C NMR

(CDCl3, δ ppm): 197.78 (Carboxylic C=O), 168.56 (Ester C=O), 139.32 (C3, C5), 134.37

(C2), 127.13 ( C6), 120.62 (C4), 114.17 (C1), 66.09 (Ester OCH3), 55.41 (Ar-OCH3),

35.04 (Ar-CH2), 29.88 (Ar-CH3); MS (70eV): m/z (%); 268 [M]+.

(32), 251 (51), 224

(65), 165 (100), 45 (25); Anal. calcd for C13H16O6: C, 58.20 H, 5.97 O, 35.82 Found: C,

58.04 H, 5.76 O, 35.59.

2.14 6-(carboxymethyl)-2,4-dimethoxy-3-methylbenzoic acid (12)

2,4-Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid (11) (8.4g,

31.3mmol) was dissolved in ethanol (75ml) and treated with KOH (5%, 125ml). The

reaction mixture was refluxed for 1h and the ethanol was rotary evaporated. The aqueous

layer was acidified with dilute hydrochloric acid to afford 6-(carboxymethyl)-2,4-

dimethoxy-3-methylbenzoic acid (12) (6.95g, 87.31%); Rf: 0.4 (petroleum ether and ethyl

acetate, 4:1); m. p. 180-182 °C; IR (KBr): 3195 (O-H), 3013 (C-H), 1741 (C=O), 1587

(C=C) cm-1

: 1H NMR (CDCl3, δ ppm): 10.91(1H, s, Ar-COOH), 10.70 (1H, s, CH2-

COOH), 7.60 (1H, s, H-6), 3.85 (3H, s, 3-OCH3), 3.67 (3H, s, 5-OCH3), 2.53 (2H, s,

CH2), 2.25 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 205.37 (Ar-COOH-C=O), 171.14

(CH2-C=O), 136.25 (C3, C5), 135.25 (C2), 133.07 (C6), 125.04 (C4), 124.41 (C1), 55.45

(2OCH3), 39.73 (Ar-CH2), 29.51 (Ar-CH3); MS (70eV): m/z (%); 254 [M]+.

(46), 237

(57), 210 (34), 165 (100); Anal. calcd for C12H14O6: C, 57.14 H, 5.55 O, 38.09 Found: C,

56.94 H, 5.36 O, 37.92.

41

2.15 6,8-dimethoxy-7-methyl-1H-isochromene-1,3(4H)-dione (13)

A solution of 6-(carboxymethyl)-2,4-dimethoxy-3-methylbenzoic acid (12) (2.5g,

9.84mmol) in dry toluene (35ml) was treated with acetic anhydride (1.1g, 10ml,

10.8mmol). The reaction mixture was refluxed for 1h and then added onto ice cold water.

The organic layer was separated, dried over anhydrous sodium sulfate and toluene was

rotary evaporated to get 6,8-dimethoxy-7-methyl-1H-isochromene-1,3(4H)-dione (13)

(1.9g, 81.89%); Rf: 0.7 (petroleum ether and ethyl acetate, 4:1); m. p. 135-136 °C; IR

(KBr): 3011 (C-H), 1735 (C=O), 1590 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 6.72 (1H,

s, H-5), 3.85 (6H, s, 2OCH3), 3.45 (2H, s, H-4), 2.28 (3H, s, Ar-CH3); 13

C NMR (CDCl3,

δ ppm): 149.57 (C1), 168.14 (C3), 163.25 (C6, C8), 136.25 (C4a), 110.51 (C7), 106.04

(C8a), 104.41 (C5), 56.35 (2OCH3), 38.73 (C4), 27.41 (Ar-CH3); MS (70eV): m/z (%);

236 [M]+.

(23), 208 (57), 192 (100), 164 (19); Anal. calcd for C12H12O5: C, 61.01 H, 5.08

O, 33.89 Found: C, 60.91 H, 4.99 O, 33.82.

2.16 General procedure for 6,8-dimethoxy-7-methyl-3-alkyl/arylisocoumarins

(16a-j)

A mixture of aliphatic/aromatic carboxylic acids (14a-j) (1mmol) and thionyl

chloride (1.2mmol) was refluxed for 1hr in the presence of a drop of DMF. The

completion of reaction was determined by stoppage of evolution of gas. Excess of the

thionyl chloride was rotary evaporated to afford acid chlorides (15a-j).

A solution of homophthalic acid anhydride (13) (2.00 mmol) in acetonitril (12ml)

was added to a solution of N, N, N’, N’-tetramethylguanidine (TMG) (2.20 mmol) in

acetonitril (5ml) over 36 min. maintaining an internal temperature of 0°C. Triethylmine

(4.0 mmol) was added in one portion. Acid chlorides (15a-j) (3.20 mmol) were added

over 3 min. and the mixture was stirred an additional 18 min. After the completion of

reaction the cooling bath was removed and reaction was allowed to warm to room

temperature. The reaction mixture was quenched by the addition of HCl (1M, 5ml). The

two phases were separated, and the organic layer was washed with saturated sodium

chloride solution and then dried (Na2SO4) prior to removal of solvent under reduced

pressure to dryness. Isocoumarins (16a-j) were then purified by preparative thin layer

chromatography using (petroleum ether and ethyl acetate, 7:3) as eluant.

42

2.17 3-Propyl-6,8-dimethoxy-7-methylisocoumarin (16a): Yield: 72%; Oil; IR

(KBr): 3031 (C-H), 1713 (C=O), 1572 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.8 (1H, s,

H-5), 7.3 (1H, s, H-4), 3.9 (6H, s, 6-OCH3,8-OCH3), 2.6 (3H, s, Ar-CH3), 2.5 (2H, t,

J=3.9, H-1’), 1.2 (2H, m, H-2’), 0.9 (3H, t, J=7.5, H-3’); 13

C NMR (CDCl3, δ ppm):

167.8 (C1), 151.2 (C3), 145.4 (C6, C8), 133.1 (C4a), 128.8 (C8a), 118.3 (C7), 110 (C4),

104.5 (C5), 53.6 (2OCH3), 38.6 (C1’), 29.7 (Ar-CH3), 21.1 (C2’), 14.3 (C3’); MS

(70eV): m/z (%); 262 [M]+.

(26), 191 (100), 71 (45), 43 (59); Anal. calcd for C15H18O4:

C, 68.70 H, 6.87; Found: C, 68.57 H, 6.69.

2.18 3-Pentyl-6,8-dimethoxy-7-methylisocoumarin (16b): Yield: 75%; Oil; IR

(KBr): 3037 (C-H), 1731 (C=O), 1559 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.9 (1H, s,

H-5), 7.3 (1H, s, H-4), 3.6 (6H, s, 6-OCH3,8-OCH3), 2.6 (3H, s, Ar-CH3), 2.3 (2H, t,

J=7.9, H-1’), 1.2-1.4 (6H, m, H-2’,H-3’,H-4’), 0.9 (3H, t, J=6.9, H-5’); 13

C NMR

(CDCl3, δ ppm): 167.8 (C1), 149.6 (C3), 141.6 (C6, C8), 133.5 (C4a), 130.9 (C8a), 118.3

(C7), 109.2 (C4), 105.3 (C5), 53.6 (2OCH3), 38.7 (C1’), 29.1 (Ar-CH3), 25.3 (C2’), 19.3

(C3’), 15.6 (C4’), 10.2 (C-5’); MS (70eV): m/z (%); 290 [M]+.

(36), 191 (100), 99 (49),

71 (34); Anal. calcd for C17H22O4: C, 70.34 H, 7.58; Found: C, 70.19 H, 7.42.

2.19 3-Heptyl-6,8-dimethoxy-7-methylisocoumarin (16c): Yield: 79%; m. p. 88-90

ºC; IR (KBr): 3021 (C-H), 1719 (C=O), 1569 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.3

(1H, s, H-5), 6.8 (1H, s, H-4), 3.9 (6H, s, 6-OCH3,8-OCH3), 2.5 (3H, s, Ar-CH3), 1.4 (2H,

t, J=6.9, H-1’), 1.2-1.3 (10H, m, H-2’,H-3’,H-4’,H-5’,H-6’), 0.9 (3H, t, J=7.2, H-7’); 13

C

NMR (CDCl3, δ ppm): 163.7 (C1), 152.9 (C3), 143.8 (C6, C8), 133.2 (C4a), 132.7 (C8a),

122.8 (C7), 114.7 (C4), 104.7 (C5), 56.2 (2OCH3), 38.7 (C1’), 29.1 (Ar-CH3), 27.8 (C2’),

21.7 (C3’), 16.8 (C4’), 11.7 (C-5’), 10.8 (C6’), 9.7 (C7’); MS (70eV): m/z (%); 318 [M]+.

(46), 191 (100), 127 (52), 99 (23); Anal. calcd for C19H25O4: C, 71.69 H, 8.17; Found: C,

71.54 H, 8.01.

2.20 3-Chloromethyl-6,8-dimethoxy-7-methylisocoumarin (16d): Yield: 70%; Oil;

IR (KBr): 3033 (C-H), 1722 (C=O), 1571 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.8

(1H, s, H-5), 6.4 (1H, s, H-4), 3.7 (6H, s, 6-OCH3,8-OCH3), 5.2 (2H, s, H-1’), 2.3 (3H, s,

Ar-CH3); 13

C NMR (CDCl3, δ ppm): 172.1 (C1), 158.3 (C3), 147.6 (C6, C8), 132.3

(C4a), 131.3 (C8a), 127.9 (C7), 114.7 (C4), 104.7 (C5), 60.0 (2OCH3), 39.6 (C1’), 30.6

43

(Ar-CH3); MS (70eV): m/z (%); 268.5 [M]+.

(36), 270.5 [M+2] (27), 191 (100), 77.5 (49),

49 (34); Anal. calcd for C13H13O4Cl: C, 58.10 H, 4.84; Found: C, 57.95 H, 4.69.

2.21 3-Hydroxymethyl-6,8-dimethoxy-7-methylisocoumarin (16e): Yield: 81%; Oil;

IR (KBr): 3345 (O-H), 3023 (C-H), 1729 (C=O), 1554 (C=C) cm-1

; 1H NMR (CDCl3, δ

ppm): 8.1 (2H, d, J=7.5, H-2’,H-6’), 8.0 (1H, s, H-5), 7.8 (7.6, dd, J=7.5), 7.4 (2H, dd,

J=7.8), 6.9 (1H, s, H-4), 3.9 (6H, s, 6-OCH3,8-OCH3), 2.6 (3H, s, Ar-CH3); 13

C NMR

(CDCl3, δ ppm): 168.5 (C1), 151.2 (C3), 141.2 (C6, C8), 131.5 (C4a), 127.8 (C8a), 118.3

(C7), 112.3 (C4), 107.3 (C5), 55.3 (2OCH3), 45.9 (C1’), 28.6 (Ar-CH3); MS (70eV): m/z

(%); 250 [M]+.

(57), 191 (100), 59 (65), 31 (54); Anal. calcd for C13H14O5: C, 62.40 H,

5.60; Found: C, 62.27 H, 5.42.

2.22 3-Phenyl-6,8-dimethoxy-7-methylisocoumarin (16f): Yield: 89%; m. p. 109-

111 oC; IR (KBr): 3033 (C-H), 1715 (C=O), 1571 (C=C) cm

-1; 1

H NMR (CDCl3, δ ppm):

8.1 (2H, d, J=7.5, H-2’,H-6’), 8.0 (1H, s, H-5), 7.6 (1H, dd, J=7.5, H-4’), 7.4 (2H, dd,

J=7.8, H-3’,H-5’), 6.9 (1H, s, H-4), 3.9 (6H, s, 6-OCH3,8-OCH3), 2.6 (3H, s, Ar-CH3);

13C NMR (CDCl3, δ ppm): 172.7 (C1), 143.7 (C3), 138.7 (C6, C8), 133.9 (C4a), 132.6

(C8a), 130.6 (C1’), 128.5 (C3’,C5’), 127.7 (C4’), 125.2 (C2’,C6’), 118.7 (C7), 112.3

(C4), 107.3 (C5), 60.8 (2OCH3), 29.7 (Ar-CH3); MS (70eV): m/z (%); 296.5 [M]+.

(63),

191 (100), 105 (51), 77 (43); Anal. calcd for C18H16O4: C, 72.79 H, 5.40; Found: C, 72.83

H, 5.26.

2.23 3-(2-chlorophenyl)-6,8-dimethoxy-7-methylisocoumarin (16g): Yield: 84%; m.

p. 119-121 o

C; IR (KBr): 3017 (C-H), 1727 (C=O), 1561 (C=C) cm-1

; 1H NMR (CDCl3, δ

ppm): 7.9 (1H, d, J=6.9, H-3’), 7.7 (1H, dd, J=3.5, H-4’), 7.6 (1H, d, J=5.5, H-6’), 7.5

(1H, dd, J=3.3, H-5’), 7.4 (1H, s, H-5), 6.8 (1H, s, H-4), 3.8 (6H, s, 6-OCH3,8-OCH3),

2.5 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 158.2 (C1), 140.4 (C3), 136.2 (C6, C8),

134.1 (C4a), 133.5 (C8a), 132.5 (C2’), 130.2 (C1’), 129.8 (C3’), 126.3 (C4’), 125.8

(C6’), 125.1 (C5’), 118.3 (C7), 114.6 (C4), 103.9 (C5), 67.3 (2OCH3), 30.2 (Ar-CH3);

MS (70eV): m/z (%); 330.5[M]+.

(59), 332.5 []M+2] (44), 191 (100), 139.5 (71), 111.5

(48); Anal. calcd for C18H15O4Cl: C, 65.35 H, 4.53; Found: C, 65.19 H, 4.39.

2.24 3-(4-methoxyphenyl)-6,8-dimethoxy-7-methylisocoumarin (16h): Yield: 87%;

m. p. 154-156 oC; IR (KBr): 3023 (C-H), 1723 (C=O), 1567 (C=C) cm

-1;

1H NMR

(CDCl3, δ ppm): 8.0 (2H, d, J=9.0, H-3’,H-5’), 7.8 (1H, s, H-5), 7.0 (1H, d, J=8.7, H-

44

2’,H-6’), 6.8 (1H, s, H-4), 3.9 (6H, s, 6-OCH3,8-OCH3), 3.8 (3H, s, 4’-OCH3), 2.6 (3H, s,

Ar-CH3); 13

C NMR (CDCl3, δ ppm): 164.0 (C1), 139.5 (C3), 137.6 (C6, C8), 134.7

(C4a), 133.8 (C8a), 131.5 (C4’), 130.2 (C1’), 126.4 (C3’,C5’), 123.8 (C2’,C6’), 121.5

(C7), 119.2 (C4), 109.1 (C5), 55.5 (6-OCH3,8-OCH3), 53.7 (4’-OCH3), 29.2 (Ar-CH3);

MS (70eV): m/z (%); 326 [M]+.

(57), 191 (100), 135 (67), 107 (47); Anal. calcd for

C19H18O5: C, 69.93 H, 5.52; Found: C, 69.74 H, 5.36.

2.25 3-(3,4-dimethoxyphenyl)-6,8-dimethoxy-7-methylisocoumarin (16i): Yield:

85%; m. p. 122-124 o

C; IR (KBr): 3029 (C-H), 1736 (C=O), 1573 (C=C) cm-1

; 1

H NMR

(CDCl3, δ ppm): 7.8 (1H, s, H-2’), 7.7 (1H, d, J=3.3, H-5’), 7.5 (1H, d, J=3.3, H-6’), 7.4

(1H, s, H-5), 7.3 (1H, s, H-4), 3.9 (6H, s, 6-OCH3,8-OCH3), 3.8 (6H, s, 3’-OCH3, 4’-

OCH3), 2.5 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 167.7 (C1), 141.1 (C3), 136.6

(C6, C8), 134.5 (C4a), 133.1 (C8a), 131.1 (C3’,C4’), 130.5 (C1’), 128.94 (C2’), 127.2

(C5’), 124.5 (C6’), 119.5 (C7), 114.8 (C4), 103.7 (C5), 68.1 (6-OCH3,8-OCH3), 56.6 (3’-

OCH3,4’-OCH3), 29.7 (Ar-CH3); MS (70eV): m/z (%); 356 [M]+.

(64), 191 (100), 165

(58), 137 (44); Anal. calcd for C19H18O5: C, 67.41 H, 5.61; Found: C, 67.28 H, 5.45.

2.26 3-(3,4,5-Trimethoxyphenyl)-6,8-dimethoxy-7-methylisocoumarin (16j): Yield:

87%; m. p. 135-137 oC; IR (KBr): 3013 (C-H), 1713 (C=O), 1587 (C=C) cm

-1;

1H NMR

(CDCl3, δ ppm): 7.5 (2H, s, H-2’,H-6’), 7.4 (1H, s, H-5), 6.9 (1H, s, H-4), 3.9 (6H, s, 6-

OCH3,8-OCH3), 3.8 (9H, s, 3’-OCH3, 4’-OCH3, 5’-OCH3), 2.5 (3H, s, Ar-CH3); 13

C

NMR (CDCl3, δ ppm): 167.8 (C1), 140.3 (C3), 135.6 (C6, C8), 134.5 (C4a), 133.1 (C8a),

132.4 (C3’,C4’,C5’), 129.6 (C1’), 122.5 (C2’,C6’), 119.5 (C7), 116.9 (C4), 107.1 (C5),

68.1 (6-OCH3,8-OCH3), 55.5 (3’-OCH3,4’-OCH3,5’-OCH3), 29.3 (Ar-CH3); MS (70eV):

m/z (%); 386 [M]+.

(79), 195 (57), 191 (100), 167 (51); Anal. calcd for C21H22O7: C,

65.28 H, 5.69; Found: C, 65.14 H, 5.57.

2.27 General procedure for 2,4-dimethoxy-3-methyl-6-(2-oxoalkyl/aryl)benzoic

acid (17a-j) A stirred solution of 6,8-Dimethoxy-7-methyl-3-alkyl/aryl Isocoumarins

(16a-j) (1.42mmol) in ethanol (20 mL) was treated with 5% KOH (40 mL) and the

mixture refluxed for four hrs. After cooling the reaction mixture, most of the ethanol was

evaporated under reduced pressure. Cold water (20 mL) was added and the mixture

acidified with dilute hydrochloric acid when solid was precipitated. Filtration followed by

drying under vacuum afforded (17a-j).

45

2.28 2,4-Dimethoxy-3-methyl-6-(2-oxopentyl)benzoic acid (17a): Yield: 78%; m. p.

133-134 oC; IR (KBr): 3224 (O-H), 3029 (C-H), 1749 (Carboxylic C=O), 1713 (Carbonyl

C=O), 1572 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 10.2 (1H, s, COOH), 7.9 (1H, s, H-

5), 4.3 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.7 (3H, s, 4-OCH3), 3.5 (2H, t, J=3.9 H-3’),

3.1 (3H, s, Ar-CH3), 1.5 (2H, m, H-4’), 0.9 (3H, t, J=7.5, H-5’); 13

C NMR (CDCl3, δ

ppm): 195.2 (C2’), 168.4 (COOH), 145.4 (C2), 139.2 (C4), 131.1 (C1), 125.1 (C6), 121.1

(C3), 108.7 (C5), 61.2 (2-OCH3), 56.4 (4-OCH3), 47.9 (C1’), 44.1 (C3’), 32.3 (Ar-CH3),

19.8 (C4’), 10.5 (C5’); MS (70eV): m/z (%); 280 [M]+ (19), 262 (43), 236 (54), 219

(100); Anal. calcd for C15H20O5: C, 64.28 H, 7.14; Found: C, 64.17 H, 7.05.

2.29 2,4-dimethoxy-3-methyl-6-(2-oxoheptyl)benzoic acid (17b): Yield: 79%; m. p.

139-140 oC; IR (KBr): 3256 (O-H), 3036 (C-H), 1753 (Carboxylic C=O), 1709 (Carbonyl

C=O), 1559 (C=C) cm-1

; 1

H NMR (CDCl3, δ ppm): 9.7 (1H, s, COOH), 8.0 (1H, s, H-5),

4.5 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.8 (3H, s, 4-OCH3), 3.7 (2H, t, J=3.7 H-3’), 3.2

(3H, s, Ar-CH3), 1.2-1.5 (6H, m, H-4’,H-5’,H-6’), 0.9 (3H, t, J=6.7, H-7’); 13

C NMR

(CDCl3, δ ppm): 197.1 (C2’), 169.2 (COOH), 143.1 (C2), 138.1 (C4), 135.2 (C1), 127.7

(C6), 119.8 (C3), 105.2 (C5), 59.5 (2-OCH3), 55.1 (4-OCH3), 49.2 (C1’), 45.5 (C3’), 30.5

(Ar-CH3), 21.5 (C4’), 18.7 (C5’), 15.7 (C6’), 10.2 (C7’); MS (70eV): m/z (%); 308 [M]+

(25), 290 (51), 264 (64) 219 (100); Anal. calcd for C17H24O5: C, 67.10 H, 7.79; Found: C,

67.02 H, 7.71.

2.30 2,4-Dimethoxy-3-methyl-6-(2-oxononyl)benzoic acid (17c): Yield: 81%; m. p.

147-148 oC; IR (KBr): 3267 (O-H), 3031 (C-H), 1743 (Carboxylic C=O), 1719 (Carbonyl

C=O), 1569 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 10.5 (1H, s, COOH), 8.2 (1H, s, H-

5), 4.1 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.8 (3H, s, 4-OCH3), 3.6 (2H, t, J=3.7 H-3’),

3.1 (3H, s, Ar-CH3), 1.3-1.6 (10H, m, H-4’,H-5’,H-6’,H-7’,H-8’), 0.9 (3H, t, J=7.3, H-

9’); 13

C NMR (CDCl3, δ ppm): 190.9 (C2’), 165.4 (COOH), 141.5 (C2), 137.1 (C4),

134.5 (C1), 125.3 (C6), 119.8 (C3), 107.4 (C5), 60.3 (2-OCH3), 52.9 (4-OCH3), 46.1

(C1’), 42.7 (C3’), 29.7 (Ar-CH3), 19.2 (C4’), 16.1 (C5’), 13.5 (C6’), 12.9 (C7’), 12.5

(C8’), 10.5 (C9’); MS (70eV): m/z (%); 336 [M]+ (25), 318 (31), 292 (51) 219 (100);

Anal. calcd for C19H28O5: C, 67.85 H, 8.33; Found: C, 67.76 H, 8.19.

2.31 6-(3-Chloro-2-oxopropyl)-2,4-dimethoxy-3-methylbenzoic acid (17d): Yield:

72%; m. p. 120-121 oC; IR (KBr): 3245 (O-H), 3033 (C-H), 1759 (Carboxylic C=O),

46

1715 (Carbonyl C=O), 1571 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 10.8 (1H, s, COOH),

8.2 (1H, s, H-5), 5.2 (2H, s, H-3’), 4.7 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.7 (3H, s, 4-

OCH3), 2.9 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 195.7 (C2’), 169.2 (COOH),

143.5 (C2), 139.2 (C4), 135.2 (C1), 132.5 (C6), 123.0 (C3), 110.6 (C5), 61.7 (2-OCH3),

56.9 (4-OCH3), 51.2 (C3’), 43.5 (C1’), 28.1 (Ar-CH3); MS (70eV): m/z (%); 286.5 [M]+

(16), 288.5 [M+2] (12), 268.5 (41), 242.5 (56), 219 (100), 49.5 (17); Anal. calcd for

C13H15O5Cl: C, 54.45 H, 5.23; Found: C, 54.37 H, 5.15.

2.32 6-(3-Hydroxy-2-oxopropyl)-2,4-dimethoxy-3-methylbenzoic acid (17e): Yield:

75%; m. p. 128-129 oC; IR (KBr): 3256 (O-H), 3030 (C-H), 1747 (Carboxylic C=O),

1721 (Carbonyl C=O), 1554 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 10.9 (1H, s, COOH),

7.6 (1H, s, H-5), 5.9 (2H, s, H-3’), 4.6 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.7 (3H, s, 4-

OCH3), 2.7 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 201.2 (C2’), 168.5 (COOH),

146.7 (C2), 137.5 (C4), 133.1 (C1), 131.8 (C6), 119.5 (C3), 109.5 (C5), 71.5 (C3’), 58.5

(2-OCH3), 55.1 (4-OCH3), 47.9 (C1’), 31.0 (Ar-CH3); MS (70eV): m/z (%); 268 [M]+

(45), 250 (57), 224 (61) 219 (100); Anal. calcd for C13H16O6: C, 58.20 H, 5.97; Found: C,

58.07 H, 5.89.

2.33 2,4-Dimethoxy-3-methyl-6-(2-oxo-2-phenylethyl)benzoic acid (17f): Yield: 89%;

m. p. 169-171 oC; IR (KBr): 3224 (O-H), 3037 (C-H), 1758 (Carboxylic C=O), 1715

(Carbonyl C=O), 1571 (C=C) cm-1

; 1

H NMR (CDCl3, δ ppm): 10.9 (1H, s, COOH), 8.3

(2H, d, J=7.5, H-2’’,H-6’’), 7.9 (1H, s, H-5), 7.7 (1H, dd, J=7.2, H-4’’), 7.5 (2H, dd,

J=7.5, H-3’’,H-5’’), 4.3 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.8 (3H, s, 4-OCH3), 2.5 (3H,

s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 194.2 (C2’), 167.5 (COOH), 143.6 (C1), 138.7

(C2), 137.6 (C1’’), 135.9 (C4), 131.7 (C6), 126.2 (C2’’,C6’’), 123.5 (C3’’,C5’’), 122.7

(C4’’), 121.3 (C3), 105.3 (C5), 62.5 (2-OCH3), 57.3 (4-OCH3), 45.3 (C1’), 29.2 (Ar-

CH3); MS (70eV): m/z (%); 314 [M]+ (39), 296 (51), 270 (65) 219 (100); Anal. calcd for

C18H18O5: C, 68.78 H, 5.73; Found: C, 65.69 H, 5.65.

2.34 6-[2-(2-Chlorophenyl)-2-oxoethyl]-2,4-dimethoxy-3-methylbenzoic acid (17g):

Yield: 84%; m. p. 175-176 oC; IR (KBr): 3228 (O-H), 3027 (C-H), 1741 (Carboxylic

C=O), 1720 (Carbonyl C=O), 1561 (C=C) cm-1

; 1

H NMR (CDCl3, δ ppm): 11.1 (1H, s,

COOH), 8.0 (1H, d, J=7.5, H-3’’), 7.7 (1H, dd, J=6.7, H-4’’), 7.6 (1H, s, H-5), 7.5 (1H,

d, J=7.2, H-6’’), 7.3 (1H, dd, J=6.9, H-5’’), 4.6 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.7

47

(3H, s, 4-OCH3), 2.7 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 201.3 (C2’), 168.1

(COOH), 145.7 (C1), 139.3 (C2), 138.2 (C2’’), 137.3 (C1’’), 136.7 (C4), 130.2 (C6),

127.7 (C3’’), 126.9 (C6’’), 124.3 (C4’’), 123.6 (C5’’), 119.2 (C3), 107.2 (C5), 61.4 (2-

OCH3), 56.7 (4-OCH3), 46.8 (C1’), 30.3 (Ar-CH3); MS (70eV): m/z (%); 348.5 [M]+

(35), 350.5 [M+2] (26), 330.5 (71), 304.5 (48), 219 (100); Anal. calcd for C18H17O5Cl: C,

61.98 H, 4.87; Found: C, 61.89 H, 4.79.

2.35 2,4-Dimethoxy-6-[2-(4-methoxyphenyl)-2-oxoethyl]-3-methylbenzoic acid (17h):

Yield: 87%; m. p. 179-180 oC; IR (KBr): 3247 (O-H), 3035 (C-H), 1761 (Carboxylic

C=O), 1723 (Carbonyl C=O), 1567 (C=C) cm-1

; 1

H NMR (CDCl3, δ ppm): 10.7 (1H, s,

COOH), 8.1 (2H, d, J=7.7, H-3’’,H-5’’), 7.9 (1H, s, H-5), 7.6 (2H, d, J=7.2, H-2’’,H-6’’),

4.3 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.8 (3H, s, 4-OCH3), 2.7 (3H, s, Ar-CH3); 13

C

NMR (CDCl3, δ ppm): 193.5 (C2’), 165.7 (COOH), 143.5 (C1), 140.7 (C2), 135.2 (C1’’),

133.3 (C4), 129.7 (C2’’,C6’’), 124.3 (C4’’), 120.3 (C3), 119.5 (C3’’,C5’’), 120.3 (C3),

104.3 (C5), 60.5 (2-OCH3), 58.5 (4’’-OCH3), 55.9 (4-OCH3), 44.7 (C1’), 28.5 (Ar-CH3);

MS (70eV): m/z (%); 344 [M]+ (49), 326 (57), 330 (69), 219 (100); Anal. calcd for

C19H20O6: C, 66.27 H, 5.81; Found: C, 66.19 H, 5.74.

2.36 2,4-Dimethoxy-6-[2-(3,4-dimethoxyphenyl)-2-oxoethyl]-3-methylbenzoic

acid (17i): Yield: 85%; m. p. 182-183 oC; IR (KBr): 3239 (O-H), 3035 (C-H), 1749

(Carboxylic C=O), 1716 (Carbonyl C=O), 1573 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm):

10.9 (1H, s, COOH), 8.0 (1H, s, H-5), 7.6 (1H, s, H-2’’), 7.5 (1H, d, J=7.1, H-5’’), 7.3

(1H, d, J=6.8, H-6’’), 4.5 (2H, s, H-1’), 3.9 (3H, s, 2-OCH3), 3.8 (3H, s, 4-OCH3), 3.7

(6H, s, 3’’-OCH3, 4’’-OCH3), 2.9 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 195.4

(C2’), 167.5 (COOH), 140.3 (C1), 139.2 (C2), 137.5 (C4), 136.6 (C1’’), 130.3 (C6’’),

128.9 (C2’’), 128.2 (C6), 125.5 (C4’’), 124.3 (C3’’), 120.4 (C3), 106.5 (C5), 61.3 (2-

OCH3), 56.2 (4-OCH3), 55.9 (3’’-OCH3), 55.6 (4’’-OCH3), 43.5 (C1’), 29.1 (Ar-CH3);

MS (70eV): m/z (%); 374 [M]+ (46), 356 (52), 330 (69), 219 (100); Anal. calcd for

C20H22O7: C, 64.17 H, 5.88; Found: C, 64.08 H, 5.79.

2.37 2,4-Dimethoxy-6-[2-(3,4,5-trimethoxyphenyl)-2-oxoethyl]-3-methylbenzoic

acid (17j): Yield: 87%; m. p. 185-187 oC; IR (KBr): 3259 (O-H), 3033 (C-H), 1757

(Carboxylic C=O), 1713 (Carbonyl C=O), 1587 (C=C) cm-1

; 1

H NMR (CDCl3, δ ppm):

10.7 (1H, s, COOH), 7.8 (1H, s, H-5), 7.7 (2H, s, H-2’’,H-6’’), 4.7 (2H, s, H-1’), 3.9 (3H,

48

s, 2-OCH3), 3.8 (3H, s, 4-OCH3), 3.7 (9H, s, 3’’-OCH3, 4’’-OCH3, 5’’-OCH3), 2.9 (3H,

s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 193.7 (C2’), 166.3 (COOH), 141.1 (C1), 139.6

(C2), 137.5 (C4), 136.7 (C1’’), 129.3 (C6), 127.5 (C2’’,C6’’), 126.7 (C3’’,C5’’), 123.4

(C4’’), 127.4 (C3), 109.1 (C5), 59.8 (2-OCH3), 57.3 (4-OCH3), 55.7 (3’’-OCH3), 55.6

(5’’-OCH3), 55.1 (4’’-OCH3), 46.2 (C1’), 28.3 (Ar-CH3); MS (70eV): m/z (%); 404 [M]+

(39), 386 (45), 360 (57), 219 (100); Anal. calcd for C21H24O8: C, 62.37 H, 5.94; Found:

C, 62.29 H, 5.85.

2.38 General procedure for 6,8-dimethoxy-7-methyl-3-alkyl/aryl-3,4-

dihydroisocoumarins (18a-j)

Sodium borohydride (18 mmol) was added portion wise to a stirred solution of

keto acids (17a-j) (0.66 mmol) in ethanol (25 mL) and water (75 mL). The reaction

mixture was stirred for 2h at room temperature, diluted with water (150 mL), acidified

with conc. HCl and stirred for a further 2h. It was then saturated with ammonium sulfate,

and extracted with ethyl acetate (3 x 100 mL). The layers were separated and the organic

layer was dried over anhydrous magnesium sulfate and concentrated. 6,8-dimethoxy-7-

methyl-3-alkyl/aryl dihydroisocoumarins (18a-j) were purified by preparative thin layer

chromatography using (petroleum ether and ethyl acetate 7:3) as eluent.

2.39 6,8-Dimethoxy-7-methyl-3-propyl-3,4-dihydroisocoumarins (18a): Yield:

75%; Oil; IR (KBr): 3029 (C-H), 1719 (C=O), 1582 (C=C) cm-1

; 1H NMR (CDCl3, δ

ppm): 6.8 (1H, s, H-5), 5.2 (1H, m, H-3), 3.9 (6H, s, 6-OCH3,8-OCH3), 3.4 (1H, dd,

Jgem=15.5, Jtrans=12.2 H-4), 3.1 (1H, dd, Jgem=15.5, Jcis=3.5 H-4), 2.9 (3H, s, Ar-CH3), 2.5

(2H, m, H-1’), 1.2 (2H, m, H-2’), 0.9 (3H, t, J=7.5, H-3’); 13

C NMR (CDCl3, δ ppm):

165.8 (C1), 147.4 (C6, C8), 143.1 (C4a), 138.8 (C8a), 129.2 (C5), 118.3 (C7), 81.1 (C3),

56.6 (6-OCH3,8-OCH3), 43.5 (C4), 35.6 (C1’), 28.7 (Ar-CH3), 19.1 (C2’), 11.2 (C3’);

MS (70eV): m/z (%); 264 [M]+ (49), 192 (100), 71 (35), 43 (65); Anal. calcd for

C15H20O4: C, 68.18 H, 7.57; Found: C, 68.09 H, 7.49.

2.40 6,8-Dimethoxy-7-methyl-3-pentyl-3,4-dihydroisocoumarins (18b): Yield:

79%; Oil; IR (KBr): 3017 (C-H), 1723 (C=O), 1579 (C=C) cm-1

; 1

H NMR (CDCl3, δ

ppm): 6.9 (1H, s, H-5), 4.9 (1H, m, H-3), 3.9 (6H, s, 6-OCH3,8-OCH3), 3.3 (1H, dd,

Jgem=15.8, Jtrans=12.6 H-4), 3.0 (1H, dd, Jgem=16.1, Jcis=3.7 H-4), 2.7 (3H, s, Ar-CH3), 2.4

(2H, m, H-1’), 1.2-1.5 (6H, m, H-2’,H-3’,H-4’), 0.9 (3H, t, J=5.9, H-5’); 13

C NMR

49

(CDCl3, δ ppm): 168.2 (C1), 149.7 (C6, C8), 145.5 (C4a), 139.9 (C8a), 127.3 (C5), 123.3

(C7), 79.3 (C3), 55.2 (6-OCH3,8-OCH3), 44.6 (C4), 38.3 (C1’), 29.3 (Ar-CH3), 20.2

(C2’), 15.3 (C3’), 13.3 (C4’), 10.6 (C5’); MS (70eV): m/z (%); 292 [M]+ (53), 192 (100),

99 (51), 71 (23); Anal. calcd for C17H24O4: C, 69.86 H, 8.21; Found: C, 69.79 H, 8.12.

2.41 6,8-Dimethoxy-7-methyl-3-heptyl-3,4-dihydroisocoumarins (18c): Yield: 81%;

m. p. 73-75 ºC; IR (KBr): 3025 (C-H), 1719 (C=O), 1569 (C=C) cm-1

; 1

H NMR (CDCl3,

δ ppm): 6.7 (1H, s, H-5), 5.3 (1H, m, H-3), 3.8 (6H, s, 6-OCH3,8-OCH3), 3.5 (1H, dd,

Jgem=15.3, Jtrans=11.8 H-4), 3.1 (1H, dd, Jgem=16.3, Jcis=3.7 H-4), 2.5 (3H, s, Ar-CH3), 2.4

(2H, m, H-1’), 1.1-1.5 (10H, m, H-2’,H-3’,H-4’,H-5’,H-6’), 0.9 (3H, t, J=5.9, H-7’); 13

C

NMR (CDCl3, δ ppm): 164.7 (C1), 151.4 (C6, C8), 142.9 (C4a), 136.3 (C8a), 121.8 (C5),

123.3 (C7), 83.7 (C3), 58.3 (6-OCH3,8-OCH3), 46.8 (C4), 37.7 (C1’), 30.1 (Ar-CH3),

19.5 (C2’), 13.6 (C3’), 12.8 (C4’), 12.0 (C5’), 11.2 (C6’), 9.8 (C7’); MS (70eV): m/z (%);

320 [M]+ (49), 192 (100), 127 (53), 99 (19); Anal. calcd for C19H28O4: C, 71.25 H, 8.75;

Found: C, 71.14 H, 8.66.

2.42 6,8-Dimethoxy-7-methyl-3-chloromethyl-3,4-dihydroisocoumarins (18d): Yield:

73%; Oil; IR (KBr): 3020 (C-H), 1728 (C=O), 1571 (C=C) cm-1

; 1

H NMR (CDCl3, δ

ppm): 6.9 (1H, s, H-5), 5.6 (1H, dd, Jgem=16.5, Jtrans=11.9 H-1’), 4.9 (1H, dd, Jgem=16.5,

Jcis=3.5 H-1’), 4.7 (1H, m, H-3), 3.7 (6H, s, 6-OCH3,8-OCH3), 3.5 (1H, dd, Jgem=15.9,

Jtrans=12.5 H-4), 3.1 (1H, dd, Jgem=16.2, Jcis=3.7 H-4), 2.7 (3H, s, Ar-CH3); 13

C NMR

(CDCl3, δ ppm): 169.3 (C1), 147.5 (C6, C8), 142.9 (C4a), 136.3 (C8a), 121.8 (C5), 127.8

(C7), 87.3 (C3), 55.5 (6-OCH3,8-OCH3), 45.7 (C1’), 40.1 (C4), 28.7 (Ar-CH3); MS

(70eV): m/z (%); 270.5 [M]+ (32), 272.5 [M+2] (24), 192 (100), 77.5 (54), 49.5 (37);

Anal. calcd for C13H15O4Cl: C, 57.67 H, 5.54; Found: C, 57.59 H, 5.49.

2.43 6,8-Dimethoxy-7-methyl-3-hydroxymethyl-3,4-dihydroisocoumarins (18e):

Yield: 85%; Oil; IR (KBr): 3367 (O-H), 3013 (C-H), 1723 (C=O), 1554 (C=C) cm-1

; 1

H

NMR (CDCl3, δ ppm): 6.7 (1H, s, H-5), 5.2 (1H, dd, Jgem=15.8, Jtrans=12.1 H-1’), 4.7

(1H, dd, Jgem=16.2, Jcis=3.3 H-1’), 4.7 (1H, m, H-3), 3.8 (6H, s, 6-OCH3,8-OCH3), 3.6

(1H, dd, Jgem=16.3, Jtrans=12.1 H-4), 3.2 (1H, dd, Jgem=15.8, Jcis=3.5 H-4), 2.6 (3H, s, Ar-

CH3), 2.0 (1H, s, -OH); 13

C NMR (CDCl3, δ ppm): 165.5 (C1), 146.6 (C6, C8), 140.5

(C4a), 138.8 (C8a), 128.9 (C7), 125.5 (C5), 89.4 (C3), 56.8 (6-OCH3,8-OCH3), 61.2

50

(C1’), 37.7 (C4), 29.2 (Ar-CH3); MS (70eV): m/z (%); 252 [M]+ (59), 192 (100), 59 (61),

31 (51); Anal. calcd for C13H16O5: C, 61.90 H, 6.34; Found: C, 61.81 H, 6.26.

2.44 6,8-Dimethoxy-7-methyl-3-phenyl-3,4-dihydroisocoumarins (18f): Yield:

84%; m. p. 92-94 ºC; IR (KBr): 3031 (C-H), 1714 (C=O), 1571 (C=C) cm-1

; 1

H NMR

(CDCl3, δ ppm): 7.9 (2H, d, J=5.8, H-2’,H-6’), 7.7 (2H, dd, J=7.9, H-3’,H-5’), 7.5 (1H,

dd, J=6.7, H-4’), 6.9 (1H, s, H-5), 5.4 (1H, dd, Jtrans=12.1, Jcis=3.3 H-3), 3.9 (6H, s, 6-

OCH3,8-OCH3), 3.4 (1H, dd, Jgem=15.7, Jtrans=12.1 H-4), 3.1 (1H, dd, Jgem=12.0, Jcis=3.7

H-4), 2.8 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 167.3 (C1), 147.3 (C6, C8), 142.3

(C4a), 139.1 (C1’), 137.2 (C8a), 131.9 (C7), 131.5 (C5), 130.0 (C2’,C6’), 123.3

(C3’,C5’), 119.5 (C4’), 83.5 (C3), 56.8 (6-OCH3,8-OCH3), 41.4 (C4); MS (70eV): m/z

(%); 298 [M]+ (67), 192 (100), 164 (37), 105 (54), 77 (47); Anal. calcd for C18H18O4: C,

72.48 H, 6.04; Found: C, 72.39 H, 5.96.

2.45 6,8-Dimethoxy-7-methyl-3-(2-chlorophenyl)-3,4-dihydroisocoumarins (18g):

Yield: 86%; m. p. 105-107 ºC; IR (KBr): 3019 (C-H), 1720 (C=O), 1561 (C=C) cm-1

; 1H

NMR (CDCl3, δ ppm): 7.9 (1H, d, J=7.4, H-3’), 7.8 (1H, d, J=6.4, H-6’), 7.6 (1H, dd,

J=6.1, H-4’), 7.5 (1H, dd, J=7.1, H-5’), 6.8 (1H, s, H-5), 5.2 (1H, dd, Jtrans=12.3, Jcis=3.7

H-3), 3.9 (6H, s, 6-OCH3,8-OCH3), 3.2 (1H, dd, Jgem=16.4, Jtrans=12.1 H-4), 3.0 (1H, dd,

Jgem=12.3, Jcis=3.6 H-4), 2.5 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 162.5 (C1),

145.7 (C6, C8), 145.5 (C4a), 139.6 (C1’), 137.6 (C8a), 136.7 (C2’), 132.3 (C5), 130.2

(C6’), 129.4 (C3’), 128.3 (C7), 126.6 (C4’), 124.5 (C5’), 88.2 (C3), 52.7 (6-OCH3,8-

OCH3), 44.7 (C4), 28.8 (Ar-CH3); MS (70eV): m/z (%); 332.5 [M]+ (59), 334.5 [M+2]

(44), 192 (100), 164 (41), 139.5 (63), 111.5 (48); Anal. calcd for C18H17O4Cl: C, 64.96 H,

5.11; Found: C, 64.89 H, 5.02.

2.46 6,8-Dimethoxy-7-methyl-3-(4-methoxyphenyl)-3,4-dihydroisocoumarins

(18h): Yield: 87%; m. p. 141-143 ºC; IR (KBr): 3027 (C-H), 1727 (C=O), 1567 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm): 7.8 (2H, d, J=6.4, H-3’,H-5’), 7.5 (2H, d, J=6.9, H-2’,H-

6’), 6.9 (1H, s, H-5), 5.3 (1H, dd, Jtrans=12.0, Jcis=3.5 H-3), 3.8 (6H, s, 6-OCH3,8-OCH3),

3.5 (1H, dd, Jgem=16.1, Jtrans=11.6 H-4), 3.1 (1H, dd, Jgem=11.8, Jcis=3.8 H-4), 2.8 (3H, s,

Ar-CH3); 13

C NMR (CDCl3, δ ppm): 164.3 (C1), 148.1 (C6, C8), 144.5 (C4a), 138.3

(C1’), 135.6 (C8a), 133.7 (C4’), 132.3 (C2’,C6’), 130.1 (C5), 129.7 (C3’,C5’), 129.3

(C7), 81.3 (C3), 56.6 (6-OCH3,8-OCH3), 52.2 (4’-OCH3), 41.6 (C4), 29.9 (Ar-CH3); MS

51

(70eV): m/z (%); 328 [M]+.

(52), 192 (100), 164 (37), 135 (57), 107 (37); Anal. calcd for

C19H20O5: C, 69.51 H, 6.09; Found: C, 69.43 H, 5.99.

2.47 6,8-Dimethoxy-7-methyl-3-(3,4-dimethoxyphenyl)-3,4-dihydroisocoumarins

(18i): Yield: 85%; m. p. 115-117 ºC; IR (KBr): 3022 (C-H), 1711 (C=O), 1573 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm): 7.8 (1H, s, H-2’), 7.7 (1H, d, J=6.8, H-5’), 7.6 (1H, d,

J=5.4, H-6’), 6.8 (1H, s, H-5), 5.1 (1H, dd, Jtrans=12.3, Jcis=3.7 H-3), 3.9 (6H, s, 6-

OCH3,8-OCH3), 3.7 (6H, s, 3’-OCH3,4’-OCH3), 3.4 (1H, dd, Jgem=15.6, Jtrans=11.4 H-4),

3.0 (1H, dd, Jgem=11.6, Jcis=3.5 H-4), 2.5 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm):

168.5 (C1), 144.5 (C6, C8), 140.6 (C4a), 137.5 (C1’), 136.9 (C8a), 133.7 (C3’), 133.2

(C4’), 131.3 (C5), 127.7 (C7), 124.5 (C6’), 123.4 (C2’), 120.2 (C5’), 83.4 (C3), 52.9 (6-

OCH3,8-OCH3), 51.3 (3’-OCH3,4’-OCH3), 45.5 (C4), 29.9 (Ar-CH3); MS (70eV): m/z

(%); 358 [M]+.

(58), 192 (100), 165 (51), 164 (45), 137 (24); Anal. calcd for C20H22O6: C,

67.03 H, 6.4; Found: C, 66.94 H, 6.06.

2.48 6,8-Dimethoxy-7-methyl-3-(3,4,5-trimethoxyphenyl)-3,4-dihydroisocoumarins

(18j): Yield: 82%; m. p. 126-127 ºC; IR (KBr): 3013 (C-H), 1732 (C=O), 1587 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm): 7.9 (2H, s, H-2’,H-6’), 6.9 (1H, s, H-5), 5.3 (1H, dd,

Jtrans=12.1, Jcis=3.5 H-3), 3.8 (6H, s, 6-OCH3,8-OCH3), 3.7 (9H, s, 3’-OCH3,4’-OCH3, 5’-

OCH3), 3.5 (1H, dd, Jgem=15.9, Jtrans=11.8 H-4), 3.1 (1H, dd, Jgem=12.0, Jcis=3.7 H-4), 2.7

(3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 168.5 (C1), 149.6 (C6, C8), 143.3 (C4a),

138.1 (C8a), 136.6 (C1’), 134.3 (C3’,C5’), 132.2 (C4’), 130.5 (C5), 127.9 (C2’,C6’),

127.7 (C7), 83.4 (C3), 56.8 (6-OCH3,8-OCH3), 54.5 (3’-OCH3,4’-OCH3, 5’-OCH3), 45.5

(C4), 30.3 (Ar-CH3); MS (70eV): m/z (%); 388 [M]+.

(69), 195 (39), 192 (100), 167 (25),

164 (19); Anal. calcd for C21H24O7: C, 64.94 H, 6.18; Found: C, 64.87 H, 6.09.

2.49 General procedure for 6,8-dihydroxy-7-methyl-3-alkyl/aryl-3,4-

dihydroisocoumarins (19a-j)

6,8-Dimethoxy-7-methyl-3-alkyl/aryl-3,4-dihydroisocoumarins (18a-j) (1.25

mmol) was dissolved in ethanethiol (3.5 mL) and solution was cooled on ice. Aluminium

chloride (3.8 mmol) was added as 3 portions with an interval of 30 min. After all the

aluminium chloride was added, the reaction mixture was stirred on ice for 1 h. The

reaction was quenched with water, alkalinized (10 % NaHCO3) and extracted with ethyl

acetate, added sodium chloride to enhance layer separation. The combined organic layers

52

were washed with brine once, dried over sodium sulfate and concentrated to give 6,8-

dihydroxy-7-methyl-3-alkyl/aryl-3,4-dihydroisocoumarins (19a-j).

2.50 6,8-Dihydroxy-7-methyl-3-propyl-3,4-dihydroisocoumarins (19a): Yield: 71%;

m. p. 93-95 °C; IR (KBr): 3474 (O-H), 3019 (C-H), 1723 (C=O), 1572 (C=C) cm-1

; 1H

NMR (CDCl3, δ ppm): 6.9 (1H, s, H-5), 5.0 (1H, m, H-3), 4.6 (2H, s, 6-OH,8-OH), 3.5

(1H, dd, Jgem=15.9, Jtrans=12.1 H-4), 3.0 (1H, dd, Jgem=15.8, Jcis=3.7 H-4), 2.7 (3H, s, Ar-

CH3), 2.4 (2H, m, H-1’), 1.2 (2H, m, H-2’), 0.9 (3H, t, J=7.5, H-3’); 13

C NMR (CDCl3, δ

ppm): 167.7 (C1), 149.4 (C6, C8), 143.1 (C4a), 138.8 (C8a), 129.2 (C5), 118.3 (C7), 81.6

(C3), 44.3 (C4), 37.3 (C1’), 29.4 (Ar-CH3), 18.1 (C2’), 10.2 (C3’); MS (70eV): m/z (%);

236 [M]+ (32), 164 (100), 136 (17), 71 (37), 43 (5); Anal. calcd for C13H16O4: C, 66.10 H,

6.77; Found: C, 66.02 H, 6.69.

2.51 6,8-Dihydroxy-7-methyl-3-pentyl-3,4-dihydroisocoumarins (19b): Yield: 73%;

m. p. °C; IR (KBr): 3456 (O-H), 3021 (C-H), 1719 (C=O), 1559 (C=C) cm-1

; 1

H NMR

(CDCl3, δ ppm): 7.1 (1H, s, H-5), 5.3 (1H, m, H-3), 4.9 (2H, s, 6-OH,8-OH), 3.4 (1H, dd,

Jgem=16.0, Jtrans=12.4 H-4), 3.1 (1H, dd, Jgem=16.3, Jcis=4.2 H-4), 2.5 (3H, s, Ar-CH3), 2.5

(2H, m, H-1’), 1.2-1.6 (6H, m, H-2’,H-3’,H-4’), 0.9 (3H, t, J=5.9, H-5’); 13

C NMR

(CDCl3, δ ppm): 163.7 (C1), 151.4 (C6, C8), 145.5 (C4a), 139.9 (C8a), 127.3 (C5), 123.3

(C7), 79.3 (C3), 42.5 (C4), 39.5 (C1’), 30.1 (Ar-CH3), 21.3 (C2’), 18.5 (C3’), 12.9 (C4’),

11.6 (C5’); MS (70eV): m/z (%); 264 [M]+ (53), 164 (100), 136 (21), 99 (57), 71 (20);

Anal. calcd for C15H20O4: C, 68.18 H, 7.57; Found: C, 68.11 H, 7.49.

2.52 6,8-Dihydroxy-7-methyl-3-heptyl-3,4-dihydroisocoumarins (19c): Yield: 79%;

m. p. 115-117 ºC; IR (KBr): 3467 (O-H), 3017 (C-H), 1727 (C=O), 1569 (C=C) cm-1

; 1

H

NMR (CDCl3, δ ppm): 6.9 (1H, s, H-5), 4.9 (1H, m, H-3), 4.5 (2H, s, 6-OH,8-OH), 3.6

(1H, dd, Jgem=15.7, Jtrans=11.9 H-4), 3.2 (1H, dd, Jgem=16.1, Jcis=3.9 H-4), 2.4 (3H, s, Ar-

CH3), 2.4 (2H, m, H-1’), 1.1-1.4 (10H, m, H-2’,H-3’,H-4’,H-5’,H-6’), 0.9 (3H, t, J=5.4,

H-7’); 13

C NMR (CDCl3, δ ppm): 166.9 (C1), 148.7 (C6, C8), 142.9 (C4a), 136.3 (C8a),

119.8 (C5), 123.3 (C7), 80.7 (C3), 43.8 (C4), 38.7 (C1’), 29.4 (Ar-CH3), 19.5 (C2’), 13.6

(C3’), 12.9 (C4’), 12.1 (C5’), 11.4 (C6’), 10.4 (C7’); MS (70eV): m/z (%); 292 [M]+ (39),

164 (100), 136 (5), 127 (43), 99 (43); Anal. calcd for C17H24O4: C, 69.86 H, 8.21; Found:

C, 69.78 H, 8.13.

53

2.53 6,8-Dihydroxy-7-methyl-3-chloromethyl-3,4-dihydroisocoumarins (19d):

Yield: 73%; m. p. 73-75 °C; IR (KBr): 3445 (O-H), 3010 (C-H), 1717 (C=O), 1571

(C=C) cm-1

; 1

H NMR (CDCl3, δ ppm): 6.9 (1H, s, H-5), 5.5 (1H, dd, Jgem=15.8,

Jtrans=12.1 H-1’), 5.1 (1H, dd, Jgem=15.6, Jcis=4.1 H-1’), 4.8 (1H, m, H-3), 4.7 (2H, s, 6-

OH,8-OH), 3.5 (1H, dd, Jgem=15.9, Jtrans=12.5 H-4), 3.1 (1H, dd, Jgem=16.2, Jcis=3.7 H-4),

2.7 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 168.9 (C1), 150.5 (C6, C8), 142.9 (C4a),

136.3 (C8a), 125.1 (C5), 127.8 (C7), 85.8 (C3), 48.7 (C1’), 41.8 (C4), 27.4 (Ar-CH3);

MS (70eV): m/z (%); 242.5 [M]+ (32), 244.5 [M+2] (24), 164 (100), 77.5 (49), 49.5 (31);

Anal. calcd for C11H11O4Cl: C, 54.43 H, 4.53; Found: C, 54.35 H, 4.35.

2.54 6,8-Dihydroxy-7-methyl-3-hydroxymethyl-3,4-dihydroisocoumarins (19e):

Yield: 81%; m. p. 89-90 °C; IR (KBr): 3481 (O-H), 3009 (C-H), 1720 (C=O), 1554

(C=C) cm-1

; 1

H NMR (CDCl3, δ ppm): 6.7 (1H, s, H-5), 5.4 (1H, dd, Jgem=16.3,

Jtrans=12.1 H-1’), 4.8 (1H, dd, Jgem=15.7, Jcis=3.8 H-1’), 4.6 (1H, m, H-3), 4.5 (2H, s, 6-

OH,8-OH), 3.6 (1H, dd, Jgem=16.3, Jtrans=12.1 H-4), 3.2 (1H, dd, Jgem=15.8, Jcis=3.5 H-4),

2.9 (3H, s, Ar-CH3), 2.1 (1H, s, -OH); 13

C NMR (CDCl3, δ ppm): 165.5 (C1), 149.2 (C6,

C8), 140.5 (C4a), 138.8 (C8a), 128.9 (C7), 121.5 (C5), 88.7 (C3), 65.3 (C1’), 39.1 (C4),

30.2 (Ar-CH3); MS (70eV): m/z (%); 224 [M]+ (34), 164 (100), 136 (31), 59 (51), 31

(43); Anal. calcd for C11H12O5: C, 58.92 H, 5.35; Found: C, 58.83 H, 5.26.

2.55 6,8-Dihydroxy-7-methyl-3-phenyl-3,4-dihydroisocoumarins (19f): Yield:

86%; m. p. 125-126 ºC; IR (KBr): 3477 (O-H), 3020 (C-H), 1722 (C=O), 1571 (C=C)

cm-1

; 1

H NMR (CDCl3, δ ppm): 7.9 (2H, d, J=5.8, H-2’,H-6’), 7.7 (2H, dd, J=7.9, H-

3’,H-5’), 7.5 (1H, dd, J=6.7, H-4’), 7.0 (1H, s, H-5), 5.3 (1H, dd, Jtrans=12.3, Jcis=3.8 H-

3), 4.7 (2H, s, 6-OH,8-OH), 3.3 (1H, dd, Jgem=15.7, Jtrans=12.1 H-4), 3.0 (1H, dd,

Jgem=12.0, Jcis=3.7 H-4), 2.5 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 162.3 (C1),

151.3 (C6, C8), 142.3 (C4a), 138.1 (C1’), 137.2 (C8a), 131.9 (C7), 129.4 (C2’,C6’),

126.3 (C3’,C5’), 123.5 (C5), 119.5 (C4’), 81.5 (C3), 44.6 (C4); MS (70eV): m/z (%); 270

[M]+ (41), 164 (100), 136 (51), 105 (51), 77 (23); Anal. calcd for C16H14O4: C, 71.11 H,

5.18; Found: C, 71.04 H, 5.11.

2.56 6,8-Dihydroxy-7-methyl-3-(2-chlorophenyl)-3,4-dihydroisocoumarins (19g):

Yield: 84%; m. p. 129-131 ºC; IR (KBr): 3488 (O-H), 3023 (C-H), 1720 (C=O), 1561

(C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.9 (1H, d, J=7.1, H-3’), 7.8 (1H, d, J=6.4, H-6’),

54

7.6 (1H, dd, J=6.5, H-4’), 7.5 (1H, dd, J=7.3, H-5’), 6.9 (1H, s, H-5), 5.5 (1H, dd,

Jtrans=12.1, Jcis=3.4 H-3), 4.9 (2H, s, 6-OH,8-OH), 3.4 (1H, dd, Jgem=16.1, Jtrans=12.1 H-

4), 3.0 (1H, dd, Jgem=12.3, Jcis=3.6 H-4), 2.9 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm):

166.5 (C1), 148.2 (C6, C8), 145.5 (C4a), 139.6 (C1’), 138.6 (C8a), 137.7 (C2’), 132.3

(C5), 130.2 (C6’), 129.4 (C3’), 128.3 (C7), 125.6 (C4’), 124.5 (C5’), 83.2 (C3), 40.7

(C4), 29.8 (Ar-CH3); MS (70eV): m/z (%); 304.5 [M]+ (35), 306.5 [M+2] (26), 164 (100),

136 (21), 139.5 (53), 111.5 (48); Anal. calcd for C16H13O4Cl: C, 63.05 H, 4.27; Found: C,

62.97 H, 4.18.

2.57 6,8-Dihydroxy-7-methyl-3-(4-methoxyphenyl)-3,4-dihydroisocoumarins (19h):

Yield: 83%; m. p. 161-162 ºC; IR (KBr): 3467 (O-H), 3018 (C-H), 1724 (C=O), 1567

(C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.7 (2H, d, J=6.9, H-3’,H-5’), 7.4 (2H, d, J=6.5,

H-2’,H-6’), 6.9 (1H, s, H-5), 5.2 (1H, dd, Jtrans=12.2, Jcis=3.7 H-3), 4.8 (2H, s, 6-OH,8-

OH), 3.5 (1H, dd, Jgem=15.8, Jtrans=11.6 H-4), 3.1 (1H, dd, Jgem=12.1, Jcis=3.9 H-4), 2.8

(3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 161.3 (C1), 150.1 (C6, C8), 144.5 (C4a),

138.3 (C1’), 135.6 (C8a), 133.7 (C4’), 132.3 (C2’,C6’), 131.7 (C5), 129.7 (C3’,C5’),

128.3 (C7), 83.5 (C3), 55.4 (4’-OCH3), 45.6 (C4), 29.9 (Ar-CH3); MS (70eV): m/z (%);

300 [M]+ (39), 164 (100), 136 (27), 135 (52), 107 (19); Anal. calcd for C17H16O5: C,

68.00 H, 5.33; Found: C, 67.93 H, 5.26.

2.58 6,8-Dihydroxy-7-methyl-3-(3,4-dimethoxyphenyl)-3,4-dihydroisocoumarins

(19i): Yield: 85%; m. p. 153-154 ºC; IR (KBr): 3479 (O-H), 3017 (C-H), 1719 (C=O),

1573 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.9 (1H, s, H-2’), 7.8 (1H, d, J=6.5, H-5’),

7.6 (1H, d, J=5.4, H-6’), 6.9 (1H, s, H-5), 5.3 (1H, dd, Jtrans=12.1, Jcis=3.4 H-3), 4.6 (2H,

s, 6-OH,8-OH), 3.7 (6H, s, 3’-OCH3,4’-OCH3), 3.4 (1H, dd, Jgem=15.6, Jtrans=11.4 H-4),

3.0 (1H, dd, Jgem=11.6, Jcis=3.5 H-4), 2.9 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm):

167.5 (C1), 146.5 (C6, C8), 140.6 (C4a), 137.5 (C1’), 136.9 (C8a), 133.7 (C3’), 133.2

(C4’), 131.3 (C5), 127.7 (C7), 126.5 (C6’), 123.4 (C2’), 120.2 (C5’), 80.4 (C3), 55.4 (3’-

OCH3,4’-OCH3), 41.5 (C4), 29.9 (Ar-CH3); MS (70eV): m/z (%); 330 [M]+ (44), 164

(100), 165 (48), 137 (20), 136 (25),; Anal. calcd for C18H18O6: C, 65.45 H, 5.45; Found:

C, 65.37 H, 5.36.

2.59 6,8-Dihydroxy-7-methyl-3-(3,4,5-trimethoxyphenyl)-3,4-dihydroisocoumarins

(19j): Yield: 82%; m. p. 145-147 ºC; IR (KBr): 3459 (O-H), 3010 (C-H), 1716 (C=O),

55

1587 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.8 (2H, s, H-2’,H-6’), 6.9 (1H, s, H-5), 5.1

(1H, dd, Jtrans=11.8, Jcis=3.7 H-3), 4.5 (2H, s, 6-OH,8-OH), 3.7 (9H, s, 3’-OCH3,4’-OCH3,

5’-OCH3), 3.5 (1H, dd, Jgem=15.9, Jtrans=11.8 H-4), 3.1 (1H, dd, Jgem=12.0, Jcis=3.7 H-4),

2.7 (3H, s, Ar-CH3); 13

C NMR (CDCl3, δ ppm): 166.5 (C1), 149.6 (C6, C8), 143.3 (C4a),

138.1 (C8a), 136.6 (C1’), 134.3 (C3’,C5’), 132.2 (C4’), 130.5 (C5), 127.9 (C2’,C6’),

127.7 (C7), 84.5 (C3), 54.7 (3’-OCH3,4’-OCH3, 5’-OCH3), 42.5 (C4), 27.9 (Ar-CH3); MS

(70eV): m/z (%); 360 [M]+ (32), 195 (34), 167 (155), 164 (100); Anal. calcd for

C19H20O7: C, 63.33 H, 5.55; Found: C, 63.26 H, 5.47.

56

3. RESULTS AND DISCUSSION

Total synthesis of a natural 3,4-dihydroisocoumarins (Stellatin) and those of the

structural analogues of some naturally occurring bioactive isocoumarins and 3,4-

dihydroisocoumarins viz. Hiburipyranone, Cytogenin, Montroumarin, Scorzocreticin,

Annulatomarin, Thunberginol B, Stoloniferol A) has been carried out.

3,5-Dimethoxy-4-methylhomophthalic was synthesized starting from commercially

available inexpensive 4-methylbenzoic acid (p-toluic acid). It was then converted into

3,5-dimethoxy-4-methyl homophthalic anhydride. The latter was then condensed with

various acyl and aroyl chlorides to afford 6,8-dimethoxy-7-methyl-3-alkyl/arylisoco-

umarins. The synthesized isocoumarins were then converted into corresponding 6,8-

dimethoxy-7-methyl-3-alkyl/aryl-3,4-dihydroisocoumarins via hydrolysis and reduction.

Finally, 3,4-dihydroisocoumarins were demethylated to give 6,8-dihydroxy-7-methyl-3-

alkyl/aryl-3,4-dihydroisocoumrins.

3.1 Synthesis of 3,5-dimethoxy-4-methylhomophthalic acid (12)

4-Methylbenzoic acid was converted into methyl 4-methylbenzoate (1) using

methanol in presence of conc. sulfuric acid as catalyst, which showed C=O stretching at

1742 cm-1

in IR spectrum.

Nuclear halogenation of the acid derivative (1) to methyl 3,5-dibromobenzoate (2)

was carried out by using “swamping catalyst method”. It involves the use of an excess of

anhydrous aluminum chloride or aluminum bromide as catalyst and no solvent is used.

The aluminum chloride complexes with the carbonyl group of the acid derivative, thus

suppressing the side chain halogenation. One mole of catalyst complexes with the

carbonyl, while the second mole increases the activity of attacking reagent, by producing

the highly electrophilic free Br+

ion or the ion pair Br+ AlCl3 Br

­.

The 4-methyl group in (1) is ideally situated for directing the incoming bromides

to C-3 and C-5 positions since the dibromination of methyl benzoate affords the methyl

3,5-dibromobenzoate.

Nucleophilic substitution of a pyridine solution of methyl 3,5-dibromobenzoate

(2) with sodium methoxide was carried out in presence of freshly prepared copper (I)

chloride as catalyst to afford methyl 3,5-dimethoxy-4-methylbenzoate. In this reaction

3,5-dimethoxy-4-methylbenzoic acid (3) was directly obtained due to concurrent

57

hydrolysis, which was confirmed by the presence of a broader stretching at 3212 cm-1

for

(O-H) in IR spectrum. In 1H NMR the six methoxy protons appeared as a singlet at δ 3.89

ppm and carboxyl showed a singlet at δ 8.95 ppm. 13

C NMR showed a signal for

carboxyl carbon at δ 168.6 ppm and the methoxy carbons appeared at δ 56.3 ppm.

The acid (3) was converted into its methyl ester (4) by using methanol in the

presence of conc. sulfuric acid as catalyst which showed the disappearance of a broad

band for (O-H) in IR spectrum. The methyl ester (4) was then reduced to 3,5-dimethoxy-

4-methylbenzyl alcohol (5) using sodium borohydride-methanol system refluxed in THF.

The IR showed a broad band at 3421 cm-1

for the hydroxyl group and also the

disappearance of carbonyl absorption was noted. The reduction of esters and similar

functional groups using sodium borohydride is relatively difficult to obtain and it has not

been widely used. However, the reactivity of the sodium borohydride can be enhanced by

carrying out the reaction in the presence of NaBH4-CH3OH. This methodology is simple,

safe, inexpensive, and general and the reduction of methyl esters was completed after

refluxing in THF.

The alcohol (5) was converted to 3,5-dimethoxy-4-methylbenzyl bromide (6) in

the presence of phosphorous tribromide (PBr3) and dry benzene in 84% yield. In IR

spectrum the absence of signals due to hydroxyl group was noticed. Nucleophilic

substitution of the bromide (6) by the cyanide, using potassium cyanide and ethanol

furnished the 3,5-dimethoxy-4-methylbenzyl cyanide (7) which showed the nitrile

absorption at 2284 cm-1

in IR spectrum.

The alkaline hydrolysis of the nitrile (7) was carried out using aqueous methanolic

potassium hydroxide in dioxane to afford the 3,5-dimethoxy-4-methylphenyl acetic acid

(8) in 71% yield. The IR showed a strong absorption at 1707 cm-1

for carbonyl and a

broad band at 3242 cm-1

for hydroxyl group. The phenylacetic acid (8) was then

converted into its methyl ester (9). In IR spectrum absorption for carbonyl appeared at

1734 cm-1

and disappearance of a broad band for hydroxyl group was observed. In 1H

NMR a singlet for methoxy protons of ester appeared at δ 3.45 ppm.

58

CH3

O

OH

CH3

O

OCH3

NaBH4/THF

CH3OH

(1)

CH3

O

OH

OCH3

H3CO

(3)

CH3

O

OCH3

Br

Br

(2)

CH3

O

OCH3

OCH3

H3CO

(4)

CH3

OCH3

H3COOH

(5)

i) Br2 /AlCl

3 (Anhd.)

ii) CH3OH

CH3OH / H+

CH3OH / H+NaOCH3, Pyridine

Cu2Cl

2 (Anhd.)

Reflux for 15 h under nitrogen

CH3

OCH3

H3CO Br

CH3

OCH3

H3CO O

OH

CH3

OCH3

H3CO O

OCH3

(6)

(8) (9)

CH3

OCH3

H3COCN

(7)

KCN

C2H5OH / H2O

CH3OH

H+

KOH / H2O

Dioxane / CH3OH

PBr3

Benzene (dry)

Scheme 3.1 Synthesis of methyl 3,5-dimethoxy-4-methylphenyl acetate (9)

Vilsmeier Haack formylation of the acetate (9) using phosphorus oxychloride in

N, N-dimethylformamide (DMF) afforded the methyl (2-formyl-3, 5-dimethoxy-4-methyl

phenyl)acetate (10). The IR showed very strong new carbonyl absorption at 1690 cm-1

for

aldehydic carbonyl in addition to the 1722 cm-1

peak for ester carbonyl already present.

The 1H NMR showed the singlet for aldehydic proton at δ 9.75 and the characteristic

changes in the chemical shifts of the benzylic protons. 13

C NMR showed a peak at δ

179.3 for aldehydic carbon and a peak at 162.4 for ester carbon already present. The

molecular ion peak appeared at m/z 252 and the base peak at m/z 165.

The aldehyde (10) was oxidized to 2,4-dimethoxy-6-(2-methoxy-2-oxoethyl)-3-

methylbenzoic acid (11) using sulfamic acid and sodium chlorite at 0°C in 79% yield.

The carbonyl absorption in IR shifted from 1690 cm-1

to 1715 cm-1

due to oxidation of

aldehydic function into carboxyl one. The absorption at 3265 cm-1

for (O-H) is also

59

present in IR spectrum. In 1H NMR, a singlet at δ 8.19 appeared for carboxyl proton and

downfield shift from δ 179.3 to δ 197.7 for carboxylic carbon was also observed in 13

C

NMR.

The alkaline hydrolysis of the ester acid (11) to 3,5-dimethoxy-4-methylhomo-

phthalic acid (12) was carried out by using 10% potassium hydroxide and ethanol in 87%

yield. The physical and FT-IR spectral data of the compounds (1-12) are shown in Table

3.1.

CH3

OCH3

H3CO O

OCH3CH3

OCH3

H3CO O

OCH3O

H

CH3

OCH3

H3CO O

OHO

OH

10%KOH

C2H5OH

(9)(10)

(12)

CH3

OCH3

H3CO O

OCH3O

OH

(11)

POCl3 / Freshly distilled DMF

CH3COO- Na

+

NH2SO3H / NaClO2 / 0 °C

H2O:THF:DMSO

Scheme 3.2 Synthesis of 3,5-dimethoxy-4-methyl homophthalic acid (12)

Table 3.1 Physical constants and FTIR spectral data of the compounds (1-12)

Compds m. p.

(°C) Rf

Yield

(%) υmax (cm

-1)

Ar-H Sp3 C-H C=O C=C O-H

1 34 0.7 95 3012 2904 1742 1562 -

2 82-84 0.6 58 3021 2917 1719 1559 -

3 210-212 0.4 74 3029 2931 1702 1569 3213

4 76-78 0.65 84 3019 2913 1732 1571 -

5 45-47 0.5 81 3023 2943 - 1554 3421

6 68-69 0.6 84 3013 2958 - 1571 -

7 48-49 0.5 84 3007 2924 - 1561 -

8 106-107 0.4 71 3013 2914 1707 1567 3242

9 38-40 0.7 88 3023 2925 1734 1573 -

10 51-53 0.55 84 3029 2917 1690, 1722 1545 -

11 164-166 0.4 79 3037 2928 1734, 1715 1562 3265

12 180-182 0.4 87 3013 2934 1741 1587 3195

Pet.Ether: Ethyl Acetate (4:1)

60

Table 3.2 1H and

13C NMR data of the compound (9)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 112.2

C-2 7.45, s 128.3

C-3 ----- 132.5

C-4 ----- 119.4

C-5 ----- 132.5

C-6 7.45, s 128.3

3-OCH3 3.96, s 55.3

5-OCH3 3.96, s 55.3

4-CH3 2.55, s 28.6

Ar-CH2 3.54, s 36.9

C=O ----- 168.2

Ester-OCH3 3.47, s 68.5

The conversion of 3,5-dimethoxy-4-methyl phenyl acetic acid (8) to methyl ester

(9) was confirmed in 1H NMR spectrum by the presence of a singlet at δ 3.47 ppm for

methoxy protons of ester and the disappearance of the signal for hydroxyl group. It was

also supported in 13

C NMR spectrum by the presence of a peak at δ 68.5 ppm for carbon

of the ester methoxy along with signals for two methoxy already present. The detailed 1H

and 13

C NMR data of the compound (9) are presented in Table 3.2.

The structure of the compound was further confirmed by mass spectrometry. The

molecular ion peak appeared at m/z 224 with 46% abundance which established the

formation of the ester (9). A peak at m/z 165 is the base peak formed by the elimination

of methoxy and carbon monoxide. The fragmentation pattern of the ester (9) is shown in

Fig. 1.

61

OCH3

H3CO

CH3

COOCH3

(m/z= 224, 46%)

-OCH3

(9)

+

..

+

OCH3

H3CO

CH3

O+

(m/z= 193, 43%)

-CO

OCH3

H3CO

CH3

+

(m/z= 165, 100%)

-CH2OOCH3

CH3

COOCH3

(m/z= 194, 37%)

-

OCH3

H3CO

CH3

CH3COO

.

(m/z= 59, 12%)

+

-CH2O

CH3

COOCH3

(m/z= 164, 23%)

+

CH3

O

(m/z= 133, 19%)

+

-OCH3

.

-CO CH3

(m/z= 105, 35%)

.+

Fig. 3.1 Mass fragmentation pattern of the compound (9)

Table 3.3 1H and

13C NMR data of the compound (10)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 117.5

C-2 ----- 131.8

C-3 ----- 136.7

C-4 ----- 121.3

C-5 ----- 136.7

C-6 7.96, s 126.2

3-OCH3 3.42, s 57.3

5-OCH3 3.25, s 57.3

4-CH3 2.80, s 32.4

Ar-CH2 2.92, s 39.1

C=O ----- 162.4

Ester-OCH3 3.11, s 61.3

CHO 9.75, s 179.3

62

The formylation of the ester (9) into methyl(2-formyl-3,5-dimethoxy-4-methyl

phenyl)acetate (10) was confirmed in 1H NMR spectrum by the presence of a singlet at δ

9.75 ppm for aldehyde proton. It was also supported in 13

C NMR spectrum by the

presence of a peak at δ 179.3 ppm for carbon of the formyl group along with signals for

two methoxy already present. The detailed 1H and

13C NMR data of the compound (10)

are presented in Table 3.3.

The structure of the compound was further confirmed by mass spectrometry. The

molecular ion peak appeared at m/z 252 with 25% abundance which proved the formation

of the aldehyde (10). By the removal of carbon monoxide from molecular ion a peak at

m/z 224 with percentage abundance 49% appeared. A peak at m/z 165 is the base peak

formed by the elimination of acetate radical from ion having m/z 224. The fragmentation

pattern of the aldehyde (10) is shown in Fig. 2.

OCH3

H3CO

CH3

COOCH3

O

H

(m/z= 252, 25%)

(10)

+

.

.

+

OCH3

H3CO

CH3

COOCH3

O

+

(m/z= 251, 65%)

-CO

OCH3

H3CO

CH3

COOCH3

+

(m/z= 223, 49%)

OCH3

CH3

COOCH3

H3CO

(m/z= 224, 49%)

.+

-

OCH3

H3CO

CH3

COOCH3

CHO

.

(m/z= 29, 31%)

+

-CH2O

CH3

COOCH3

H3CO

(m/z= 194, 21%)

+

CH3

COOCH3

(m/z= 164, 27%)

CH3

(m/z= 105, 31%)

-H

-CO

-CH2O

-CH3COO

OCH3

CH3

H3CO

(m/z= 165, 100%)

+

+

-CH3COO

Fig. 3.2 Mass fragmentation pattern of the compound (10)

63

Table 3.4 1H and

13C NMR data of the compound (11)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 114.1

C-2 ----- 134.3

C-3 ----- 139.3

C-4 ----- 120.6

C-5 ----- 139.3

C-6 7.66, s 127.1

3-OCH3 3.82, s 55.4

5-OCH3 3.67, s 55.4

4-CH3 2.25, s 35.0

Ar-CH2 2.92, s 39.1

C=O ----- 168.5

Ester-OCH3 3.63, s 66.0

COOH 8.19, s 197.7

OCH3

H3CO

CH3

COOCH3

O

OH

(m/z= 268, 32%)

(11)

+

.

.

+

OCH3

H3CO

CH3

COOCH3

O

+

(m/z= 251, 51%)

-CO

OCH3

H3CO

CH3

COOCH3

+

(m/z= 223, 36%)

OCH3

CH3

COOCH3

H3CO

(m/z= 224, 65%)

.+

-

OCH3

H3CO

CH3

COOCH3

COOH

.

(m/z= 45, 25%)

+

-CH2O

CH3

COOCH3

H3CO

(m/z= 194, 16%)

+

CH3

COOCH3

(m/z= 164, 32%)

CH3

(m/z= 105, 23%)

-CH2O

-CH3COO

OCH3

CH3

H3CO

(m/z= 165, 100%)

+

+

-CH3COO

-OH

-CO2

Fig. 3.3 Mass fragmentation pattern of the compound (11)

64

Table 3.5 1H and

13C NMR data of the compound (12)

Carbons

δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 124.4

C-2 ----- 135.2

C-3 ----- 136.2

C-4 ----- 125.0

C-5 ----- 136.2

C-6 7.60, s 133.0

3-OCH3 3.85, s 55.4

5-OCH3 3.67, s 55.4

4-CH3 2.25, s 29.5

Ar-CH2 2.53, s 39.7

Ar-COOH 10.91, s 205.3

COOH 10.70, s 171.1

The formation of the 3,5-dimethoxy-4-methylhomophthalic acid (12) was

confirmed by the presence of two singlets at δ 10.91 ppm and 10.70 ppm in 1H NMR

spectrum for two protons of two carboxylic acids. It was also supported by 13

C NMR

spectrum due to the presence of two peaks at δ 205.3 and 171.1 ppm for two carboxylic

groups. The detailed 1H and

13C NMR data of the compound (12) are presented in Table

3.5.

In mass spectrometry, the molecular ion peak appeared at m/z 254 with

percentage abundance 46% which confirmed the formation of homophthalic acid (12). By

the removal of water from molecular ion, a peak at m/z 236 with percentage abundance

48% appeared. A peak at m/z 192 is the base peak formed by the elimination of carbon

dioxoide from the ion having m/z 236. The fragmentation pattern of the homophthalic

acid (12) is shown in Fig. 4.

65

OCH3

H3CO

CH3

COOH

O

OH

(m/z= 254, 46%)

(12)

.

.

+

OCH3

H3CO

CH3

COOH

O

+

(m/z= 237, 57%)

-CO

OCH3

H3CO

CH3

COOH+

(m/z= 209, 23%)

OCH3

CH3

COOHH3CO

(m/z= 210, 34%)

.+

-

(m/z= 236, 48%)

CH3

CH3H3CO

OCH3

(m/z= 166, 29%)

+

CH3

CH3H3CO

(m/z= 136, 18%)

-CH2O

(m/z= 192, 100%)

+

+

-OH

-CO2

H2O

+

OCH3

H3CO

CH3

O

O

OCH2

OCH3

CH3

CH3

-CO2

-CO2

.

-CH2O

CH3

CH3

(m/z= 106, 25%)

+

Fig. 3.4 Mass fragmentation pattern of the compound (12)

3.2 Synthesis of 6,8-dimethoxy-7-methyl-3-alkyl/aryl isocoumarins (16a-j)

3,5-Dimethoxy-4-methylhomophthalic acid (12) was converted into

corresponding anhydride (13) by refluxing it with acetic anhydride in the presence of dry

toluene as solvent. The formation of the anhydride (13) was confirmed by the presence of

absorption at 1735 cm-1

and disappearance of the absorption for hydroxyl groups in IR

spectrum. The 1H NMR and

13C NMR data of the homophthalic anhydride (13) are

presented in Table 3.7.

O

O

OCH3

H3CO

CH3

OHOH

(12)

O

O

O

OCH3

H3CO

CH3

(13)

Acetic Anhydride

Dry Toluene

66

Table 3.6 Physical constants and FT-IR spectral data of the compound (13)

Compd M.P.

°C

Yield

% Rf

υmax (cm-1

)

Ar-H Sp3 C-H C=O C=C

13 135-136 82 0.7 3011 2913 1735

1590

Table 3.7 1H and

13C NMR data of the compound (13)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 149.5

C-3 ----- 168.1

C-4 3.45, s 38.7

C-4a ----- 136.2

C-5 6.72, s 104.4

C-6 ----- 163.2

C-7 ----- 110.5

C-8 ----- 163.2

C-8a ----- 106.0

6-OCH3 3.85, s 56.3

8-OCH3 3.85, s 56.3

7-CH3 2.28, s 27.4

In 1H NMR spectrum the methylene protons of C-4 appeared as a singlet at 3.45

ppm. Aromatic hydrogen H-5 gives a singlet at δ 6.72 and a singlet for two methoxy

groups appeared at δ 3.85. The structure of the compound was also confirmed by 13

C

NMR. The carbonyl carbons C1 and C3 appeared at δ 149.5 and 168.1 respectively.

The aliphatic and aromatic carboxylic acids (14a-j) were converted into their

respective acid chlorides by treatment with thionyl chloride (15a-j) in the presence of

catalytic amount of DMF. Acid chlorides (15a-j) were then condensed with homophthalic

anhydride (13) in the presence of triethyl amine and tetramethyl guanidine to afforded

6,8-dimethoxy-7-methyl-3-alkyl/arylisocoumarins (16a-j). These isocoumarins were

purified by preparative thin layer chromatography.

67

The synthesized isocoumarins (16a-j) showed the characteristic absorption for

lactonic carbonyl at 1713-1736 cm-1

in IR spectra. The physical constants and FTIR

spectral data of the compounds (16a-j) is shown in table 3.8 and the elemental analysis

data of these isocoumarins is presented in table 3.9. The isocoumarins showed the

characteristic 1H singlet of isocoumarin moiety (H-4) at δ 6.4-7.3 ppm in 1H NMR

spectrum and the lactonic carbonyl carbon in 13

C NMR showed the peak at δ 158-170

ppm.

R=

14a = 14b = 14c =

14d = -CH2Cl 14e = -CH2OH

14f = 14g = 14h = 14i = 14j =Cl

OCH3

OCH3

OCH3

OCH3

OCH3

H3CO

R

O

OH

+ SOCl2 R

O

Cl

(15a-j)

CH2 CH3_

CH2 CH3_

CH2 CH3_

(14a-j)

R is same as in (14a-j)

R

O

Cl

+O

O

O

OCH3

H3CO

CH3

(13)

O

OOCH3

H3CO

CH3

R

(16a-j)(15a-j)

TMG / (C2H5)3N

CH3CN

R is same as in (14a-j)

Scheme 3.3 Synthesis of 6,8-dimethoxy-7-methyl-3-alkyl/arylisocoumarins (16a-j)

68

Table 3.8 Physical constants and FTIR spectral data of the compounds (16a-j)

Compds. M. P.

(°C) Rf

Yield

(%) υmax (cm

-1)

Ar-C-H Sp3 C-H C=O C=C

16a oil 0.7 72 3031 2924 1713 1572

16b oil 0.8 75 3037 2933 1731 1559

16c 88-90 0.8 79 3021 2924 1719 1569

16d oil 0.6 70 3033 2931 1722 1571

16e oil 0.5 81 3023

3345(O-H) 2914 1729 1554

16f 109-111 0.6 89 3033 2931 1715 1571

16g 119-121 0.7 84 3017 2924 1727 1561

16h 154-156 0.5 87 3023 2914 1723 1567

16i 122-124 0.6 85 3029 2925 1736 1573

16j 135-137 0.7 87 3013 2934 1713 1587

Pet. Ether: Ethyl Acetate (4:1)

Table 3.9 Elemental analysis data of the compounds (16a-j)

Compounds

Molecular Formula

Calculated Found

C H C H

C15H18O4 (16a) 68.70 6.87 68.57 6.69

C17H22O4 (16b) 70.34 7.58 70.19 7.42

C19H26O4 (16c) 71.69 8.17 71.54 8.01

C13H13O4Cl (16d) 58.10 4.84 57.95 4.69

C13H14O5 (16e) 62.40 5.60 62.27 5.42

C18H16O4 (16f) 72.97 5.40 72.83 5.26

C18H15O4Cl (16g) 65.35 4.53 65.19 4.39

C19H18O5 (16h) 69.93 5.52 69.74 5.36

C20H20O6 (16i) 67.41 5.61 67.28 5.45

C21H22O7 (16j) 65.28 5.69 65.14 5.57

69

Table 3.10 1H and

13C NMR data of compound (16a)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 167.8

C-3 ----- 151.2

C-4 7.3, s 110.3

C-4a ----- 133.1

C-5 7.8, s 104.5

C-6 ----- 145.4

C-7 ----- 118.3

C-8 ----- 145.4

C-8a ----- 128.8

C-1’ 2.5, t, J=3.9 38.6

C-2’ 1.2, m 21.1

C-3’ 0.9, t, J=7.5 14.3

6-OCH3 3.9, s 53.6

8-OCH3 3.9, s 53.6

7-CH3 2.6, s 29.7

The formation of the 6,8-dimethoxy-7-methyl-3-propylisocoumarin (16a) was confirmed

by the presence of a singlet at δ 7.3 ppm in 1H NMR spectrum for H-4. It was also

supported in 13

C NMR spectrum by the presence of a peak at δ 167.8 ppm for lactonic

carbon. Table 3.10 gives the 1H and

13C NMR data of the compound (16a).

In mass spectrometry the molecular ion peak appeared at m/z 262 with 26%

abundance which confirmed the formation of isocoumarin (16a). By the removal of

propyl radical from molecular ion, a peak appeared at m/z 219 with 47% abundance. A

peak at m/z 191 is the base peak formed by the elimination of butanoyl radical from

molecular ion. The fragmentation pattern of the isocoumarin (16a) is shown in fig. 5.

The structures of all other isocoumarins (16b-j) were confirmed by the presence

H-4 singlet in 1H NMR along with other signals in acceptable regions. The signal for

lactonic carbonyl carbon in 13

C NMR also helpful for the confirmation of structures of all

these compounds.

70

(m/z = 262, 26%)(m/z = 191, 100%)

CH+

O

H3CO

OCH3

CH3

._

(m/z = 71, 45%)

(m/z = 163, 55%)

CH+

H3CO

OCH3

CH3

CO_

CO_

(m/z =43, 59%)

.

O

OCH3

CH3

H3CO

_

_ .

O

O

H3CO

OCH3

CH3

(m/z = 219, 47%)

CO2_ H3CO

OCH3

CH3

(m/z = 175, 17%)

_CO

O

OCH3

CH3

CH3H3CO

(m/z = 234, 40%)

+.

++

+

+

+

+

+

O

O

H3CO

OCH3

CH3

CH3

OCH3OCH3

CH3CH2CH2

CH3CH2CH2

.

CH3

(m/z = 115, 15%)

+-2CH2O

(16a)

Fig. 3.5 Mass fragmentation pattern of the compound (16a)

Table 3.11 1H and

13C NMR data of compound (16e)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 168.5

C-3 ----- 151.2

C-4 6.6, s 112.3

C-4a ----- 131.5

C-5 6.9, s 107.3

C-6 ----- 141.2

C-7 ----- 118.3

C-8 ----- 141.2

C-8a ----- 127.8

C-1’ 4.8, s 45.9

O-H 2.4, s ---

6-OCH3 3.8, s 53.3

8-OCH3 3.8, s 53.3

7-CH3 2.5, s 28.6

71

(m/z = 250, 57%)(m/z = 191, 100%)

CH+

O

H3CO

OCH3

CH3

._

(m/z = 233, 41%)

(m/z = 163, 55%)

CH+

H3CO

OCH3

CH3

CO_

CO_

(m/z =205, 23%)

.

_.

O

O

H3CO

OCH3

CH3

(m/z = 219, 53%)

CO2_ H3CO

OCH3

CH3

(m/z = 175, 21%)

_CO

O

OCH3

CH3

OHH3CO

(m/z = 222, 31%)

+.

++

+

++

+

+

O

O

H3CO

OCH3

CH3

OH

OOH

CH2OH

.

CH3

(m/z = 115, 17%)

+-2CH2O

(16e)

-OHO

O

H3CO

OCH3

CH3

O

H3CO

OCH3

CH3

Fig. 3.6 Mass fragmentation pattern of the compound (16e)

Table 3.12 1H and

13C NMR data of compound (16h)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 164.0

C-3 139.5

C-4 6.6, s 119.2

C-4a ----- 134.7

C-5 6.9, s 109.1

C-6 ----- 132.6

C-7 ----- 121.5

C-8 ----- 132.6

C-8a ----- 133.8

C-1’ ----- 130.2

C-2’ 7.0, d, J=8.7 123.8

C-3’ 8.0, d, J=9.0 126.4

C-4’ ----- 131.5

C-5’ 8.0, d, J=9.0 126.4

C-6’ 7.0, d, J=8.7 123.8

4’-OCH3 3.8, s 53.7

6-OCH3 3.9, s 55.5

8-OCH3 3.9, s 55.5

7-CH3 2.6, s 29.2

72

O

O

H3CO

OCH3

CH3

OCH3

(m/z = 326, 47%)(m/z = 191, 100%)

CH+

O

H3CO

OCH3

CH3

O

H3CO

.

_

O

H3CO

(m/z = 135, 57%)

(m/z = 163, 55%)

CH+

H3CO

OCH3

CH3

CO_

CO_

H3CO

m/z = 77, 23%

.

O

OCH3

CH3

H3CO

_

OCH3

_

.

O

O

H3CO

OCH3

CH3

(m/z = 219, 39%)

CO2_ H3CO

OCH3

CH3

(m/z = 175, 21%)

_CO.

O

OCH3

CH3

OCH3

(m/z = 298, 31%)

+ .

++

+

+

+

+

+

(16h)

CH3

(m/z = 115, 17%)

+-2CH2O

Fig. 3.7 Mass fragmentation pattern of the compound (16h)

3.3 Synthesis of 6,8-dimethoxy-7-methyl-3-alkyl/aryl-3,4-dihydroisocoumarins

(18a-j)

Alkaline hydrolysis of the 6,8-dimethoxy-7-methyl-3-alkyl/arylisocoumarins

(16a-j) to furnish the 2,4-dimethoxy-3-methyl-6-(2-oxoalkyl/aryl)benzoic acid (17a-j)

was accomplished in 75-85% yield. The ketonic and carboxylic carbonyl absorptions

were observed in IR spectra at 1743-1759 and 1709-1719 cm-1

, respectively. The physical

constants and the IR spectral data of the keto acids (17a-j) are given in Table 3.13 and the

elemental analysis data in Table 3.14. The keto acids (17a-j) showed the characteristic

2H singlet at δ 4.3-4.7 (H-4, Ar-CH2) in the 1H NMR spectrum. The C-1 and C-4

appeared at δ 165-169 and δ 43-47, respectively, in 13

C NMR.

73

R=

16a = 16b = 16c =

16d = -CH2Cl 16e = -CH2OH

16f = 16g = 16h = 16i = 16j =Cl

OCH 3

OCH3

OCH3

OCH3

OCH3

H3CO

CH2 CH3_

CH2 CH3_

CH2 CH3_

O

OOCH3

H3CO

CH3

R

(16a-j)

OOCH3

H3CO

CH3

R

O

OH

(17a-j)

OOCH3

H3CO

CH3

R

OHOH

H

5%KOH(aq)

C2H

5OH, 4h reflux

NaBH4

C2H

5OH, 2h R.T

OOCH3

H3CO

CH3

R

O

H

(18a-j)

R is same as in (16a-j)

R is same as in (16a-j)

Scheme 3.4 Synthesis of 6,8-dimethoxy-7-methyl-3-alkyl/ary-3,4-

dihydroisocoumarins (18a-j)

74

Table 3.13 Physical constants and FTIR spectral data of the compounds (17a-j)

Compds. M. P.

(°C) Rf

Yield

(%)

υmax (cm-1

)

Ar-

C-H

Sp3

C-H C=O C=C O-H

17a 133-134 0.4 78 3029 2914 1749, 1713 1572 3224

17b 139-140 0.45 79 3036 2931 1753, 1709 1559 3256

17c 147-148 0.5 81 3031 2914 1743, 1719 1569 3267

17d 120-121 0.4 72 3033 2933 1759, 1715 1571 3245

17e 128-129 0.35 75 3030 2924 1747, 1721 1554 3256

17f 169-171 0.3 89 3032 2935 1758, 1715 1571 3237

17g 175-176 0.35 84 3027 2926 1741, 1720 1561 3228

17h 179-180 0.3 87 3029 2919 1761, 1723 1567 3247

17i 182-183 0.3 85 3035 2927 1749, 1716 1573 3239

17j 185-187 0.3 87 3033 2915 1757, 1713 1587 3259

Pet. Ether: Ethyl Acetate (4:1)

Table 3.14 Elemental analysis data of the compounds (17a-j)

Compounds

Molecular Formula

Calculated Found

C H C H

C15H20O5 (17a) 64.28 7.14 64.17 7.05

C17H24O5 (17b) 67.10 7.79 67.02 7.71

C19H28O5 (17c) 67.85 8.33 67.76 8.19

C13H15O5Cl (17d) 54.45 5.23 54.37 5.15

C13

H16

O6 (17e) 58.20 5.97 58.07 5.89

C18H18O5 (17f) 68.78 5.73 68.69 5.65

C18H17O5Cl (17g) 61.98 4.87 61.89 4.79

C19H18O6 (17h) 66.27 5.81 66.19 5.74

C20H22O7 (17i) 64.17 5.88 64.08 5.79

C21H24O8 (17j) 62.37 5.94 62.29 5.85

75

Table 3.15 1H and

13C NMR data of compound (17a)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 131.1

C-2 ----- 145.4

C-3 ----- 121.1

C-4 ----- 139.2

C-5 7.9 s 108.7

C-6 ----- 125.1

C-1’ 4.3, s 47.9

C-2’ ----- 195.2

C-3’ 3.5, t, J=3.9 44.1

C-4’ 15, m 19.8

C-5’ 0.9, t, J=7.5 10.5

-COOH 10.2, s 168.4

2-OCH3 3.9, s 61.2

4-OCH3 3.7, s 56.4

3-CH3 3.1, s 32.3

The formation of the keto acid (17a) was confirmed by the presence of a singlet at

δ 10.2 ppm in 1H NMR spectrum for carboxyl hydrogen. It was also supported in

13C

NMR spectrum by the presence of a peak at δ 168.4 ppm for carboxylic carbon. The

detailed 1H and

13C NMR data of the compound (17a) are presented in Table 3.15.

The structure of the compound was further confirmed by mass spectrometry. The

molecular ion peak appeared at m/z 280 with 19% abundance which confirmed the

formation of isocoumarin (17a). By the removal of a water molecule from molecular ion

a peak at m/z 262 with percentage abundance 43 appeared. A peak at m/z 219 is the base

peak formed by the elimination of isopropyl radical from ion having m/z 262. The

fragmentation pattern of the keto acid (17a) is shown in Fig. 8.

The structures of all other keto acids (17b-j) were confirmed by the presence

carboxyl proton singlet in 1H NMR along with other signals in acceptable regions. The

signal for carboxylic carbon in 13

C NMR is also helpful for the confirmation of structures

of all these compounds.

76

OOCH3

H3CO

CH3

CH3

OH

O

(17a)

+.

-H2OOOCH3

H3CO

CH3

CH3

+ .

-CO2OCH3

H3CO

CH3

CH3

O

+.

CH3

O

+

-COCH3 CH3

+

OCH3

H3CO

CH3

_

OOCH3

H3CO

CH3

+

OCH3

H3CO

CH3

CH3

O

_

+

CH3

CH3.

_

H3CO

CH3

+

-OCH3

..

(m/z = 280, 19%)

.

(m/z = 236, 54%)

(m/z = 165, 61%)(m/z = 134, 31%) (m/z = 219, 100%)

(m/z = 262, 43%)

(m/z = 43, 67%)(m/z = 71, 39%)

Fig. 3.8 Mass fragmentation pattern of the compound (17a)

Table 3.16 1H and

13C NMR data of compound (17f)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 143.6

C-2 ----- 138.7

C-3 ----- 121.3

C-4 ----- 131.7

C-5 7.9, s 105.3

C-6 ----- 135.9

C-1’ ----- 137.6

C-2’ 8.3, d, J=7.5 126.2

C-3’ 7.5, dd, J=7.1, 6.9 123.5

C-4’ 7.9, dd, J=7.2, 6.7 122.7

C-5’ 7.5, dd, J=7.1, 6.9 123.5

C-6’ 8.3, d, J=7.5 126.2

-CH2 4.3, s 45.3

C=O ----- 194.2

-COOH 10.9, s 167.5

2-OCH3 3.9, s 62.5

4-OCH3 3.8, s 57.3

3-CH3 2.5, s 29.2

77

OOCH3

H3CO

CH3OH

O

(17f)

+.

-H2OOOCH3

H3CO

CH3

+.

-CO2OCH3

H3CO

CH3

O

+.

O

+

-CO

+

OCH3

H3CO

CH3

_

OOCH3

H3CO

CH3

+

OCH3

H3CO

CH3O

_

+

CH3

CH3.

_

H3CO

CH3 O

+

-OCH3

.

(m/z = 314, 39%)

.

(m/z = 270, 65%)

(m/z = 192, 79%) (m/z = 161, 19%) (m/z = 219, 100%)

(m/z = 296, 51%)

(m/z = 77, 29%)(m/z = 105, 35%)

.

Fig. 3.9 Mass fragmentation pattern of the compound (17f)

Sodium borohydride reduction of the keto acids (17a-j) afforded the

corresponding racemic hydroxy acids which underwent spontaneous cyclodehydration on

standing for a few minutes (as monitored by TLC) to afford (±)-6,8-dimethoxy-7-methyl-

3-alkyl/aryl-3,4-dihydroisocoumarins (18a-j) without any dehydrating agent. The

methylene protons (C-4) adjacent to the newly generated chiral center (C-3) in

dihydroisocoumarins (18a-j) showed the diastereotopic effect.

Each hydrogen at C-4 was split by the other nearly to same extent and unequally

by the adjacent methane proton. The double doublet of the hydrogen cis to phenyl ring is

located slightly upfield and that of trans hydrogen is located slightly downfield. The H-3

hydrogen showed the vicinal coupling to the trans and to the cis protons present in its

vicinity.

78

Table 3.17 Physical constants and FTIR spectral data of the compounds (18a-j)

Compds. M. P.

(°C) Rf

Yield

(%) υmax (cm

-1)

Ar-C-H Sp3 C-H C=O C=C

18a oil 0.7 75 3029 2933 1719 1582

18b oil 0.8 79 3017 2923 1723 1579

18c 73-75 0.8 81 3025 2924 1719 1569

18d oil 0.6 73 3020 2932 1728 1571

18e oil 0.5 85 3013

3367(O-H) 2923 1723 1554

18f 92-94 0.6 84 3031 2919 1714 1571

18g 105-107 0.7 86 3019 2921 1720 1561

18h 141-143 0.5 87 3027 2917 1727 1567

18i 115-117 0.6 85 302 2923 1711 1573

18j 126-127 0.7 82 3013 2934 1732 1587

Pet. Ether: Ethyl Acetate (4:1)

Table 3.18 Elemental analysis data of the compounds (18a-j)

Compounds

Molecular Formula

Calculated Found

C H C H

C15H18O4 (18a) 68.18 7.57 68.09 7.49

C17H22O4 (18b) 69.86 8.21 69.79 8.12

C19H26O4 (18c) 71.25 8.75 71.14 8.66

C13H13O4Cl (18d) 57.67 5.54 57.59 5.49

C13H14O5 (18e) 61.90 6.34 61.81 6.26

C18H16O4 (18f) 72.48 6.04 72.39 5.96

C18H15O4Cl (18g) 64.96 5.11 64.89 5.02

C19H18O5 (18h) 69.51 6.09 69.43 5.99

C20H20O6 (18i) 67.03 6.14 66.94 6.06

C21H22O7 (18j) 64.94 6.18 64.87 6.09

79

Table 3.19 1H and

13C NMR data of compound (18a)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 165.8

C-3 5.2, m 81.3

C-4 3.1, dd, J= 15.5, 3.5

3.4, dd, J=15.5, 12.2 43.5

C-4a ----- 143.1

C-5 6.8, s 129.2

C-6 ----- 147.4

C-7 ----- 118.3

C-8 ----- 145.4

C-8a ----- 138.8

C-1’ 2.5, m 35.6

C-2’ 1.2, m 19.1

C-3’ 0.9, t, J=7.5 11.2

6-OCH3 3.9, s 56.6

8-OCH3 3.9, s 56.6

7-CH3 2.9, s 28.7

The formation of 6,8-dimethoxy-7-methyl-3-propyl-3,4-dihydroisocoumarin

(18a) was confirmed by the presence of two double doublets at δ 3.1 and 3.4 ppm in 1H

NMR spectrum for H-4 hydrogens, a multiplet at δ 5.2 ppm for H-3 hydrogen and

disappearance of singlet for carboxylic proton. It was also supported in 13

C NMR

spectrum by the presence of a peak at δ 165.8 ppm for lactonic carbonyl carbon. The

detailed 1H and

13C NMR data of the compound (18a) are presented in Table 3.19. The

molecular ion peak appearing at m/z 264 with percentage abundance 37, confirmed the

formation of dihydroisocoumarin (18a). By the removal of a molecule of butanal from

molecular ion, a peak arose at m/z 192 (base peak). The fragmentation pattern of the

dihydroisocoumarin (18a) is shown in Fig. 10.

The detailed 1H and

13C NMR data of the dihydroisocoumarins (18e) and (18f)

are presented in Tables 3.20 and 3.21 respectively. The mass fragmentation pattern of the

(18e) and (18f) are shown in Fig. 11 and 12, respectively.

80

(m/z = 264, 37%)(m/z = 192, 100%)

O

H3CO

OCH3

CH3

CH2

_

(m/z = 72, 52%)

(m/z = 164, 55%)

H3CO

OCH3

CH3

CH2

CO_

CO_

(m/z =44, 59%)

.

O

OCH3

CH3

H3CO

_

_ .

O

O

H3CO

OCH3

CH3

(m/z = 221, 32%)

CO2_ H3CO

OCH3

CH3

(m/z = 177, 21%)

_CO

O

OCH3

CH3

CH3H3CO

(m/z = 236, 43%)

+.

++

+

+

+

+

+

O

O

H3CO

OCH3

CH3

CH3

OCH3

H

OCH3

H

CH3CH2CH3

CH3CH2CH2

.

CH3

(m/z = 117, 11%)

+-2CH2O

(18a)

.

.

.

Fig. 3.10 Mass fragmentation pattern of the compound (18a)

Table 3.20 1H and

13C NMR data of compound (18e)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 165.5

C-3 4.7, m 89.4

C-4 3.2, dd, J= 15.8, 3.5

3.6, dd, J=16.3, 12.1 37.7

C-4a ----- 140.5

C-5 6.7, s 125.5

C-6 ----- 146.6

C-7 ----- 128.9

C-8 ----- 146.6

C-8a ----- 138.6

C-1’ 5.2, dd, J=15.8, 12.1

4.7, dd, J=16.2, 3.3 61.2

O-H 2.0, s ---

6-OCH3 3.8, s 56.8

8-OCH3 3.8, s 56.8

7-CH3 2.6, s 29.2

81

(m/z = 252, 61%)(m/z = 192, 100%)

CH2

O

H3CO

OCH3

CH3

.

_

(m/z = 235, 37%)

(m/z = 164, 48%)

CH2H3CO

OCH3

CH3

CO_

CO_

(m/z =207, 29%)

.

_.

O

O

H3CO

OCH3

CH3

(m/z = 221, 57%)

CO2_ H3CO

OCH3

CH3

(m/z = 177, 17%)

_CO

O

OCH3

CH3

OHH3CO

(m/z = 224, 34%)

+.

++

+

++

+

+

O

O

H3CO

OCH3

CH3

OH

OOH

H

CH2OH

.

CH3

(m/z = 117, 23%)

+-2CH2O

(18e)

-OHO

O

H3CO

OCH3

CH3

O

H3CO

OCH3

CH3

.

Fig. 3.11 Mass fragmentation pattern of the compound (18e)

Table 3.21 1H and

13C NMR data of compound (18f)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 167.3

C-3 5.4, dd, J= 12.1, 3.3 83.5

C-4 3.1, dd, J= 12.0, 3.7

3.4, dd, J=15.7, 12.1 41.4

C-4a ----- 142.3

C-5 6.9, s 131.5

C-6 ----- 147.3

C-7 ----- 131.9

C-8 ----- 147.3

C-8a ----- 137.2

C-1’ ----- 139.1

C-2’ 7.9, d, J=5.8 130.0

C-3’ 7.7, dd, J=7.9, 6.1 123.3

C-4’ 7.5, dd, J=6.7, 5.1 119.5

C-5’ 7.7, dd, J=7.9, 6.1 123.3

C-6’ 7.9, d, J=5.8 130.0

6-OCH3 3.9, s 56.8

8-OCH3 3.9, s 56.8

7-CH3 2.8, s 30.1

82

O

O

H3CO

OCH3

CH3

(m/z = 298, 56%)(m/z = 192, 100%)

O

H3CO

OCH3

CH3

CH2

.

_

(m/z = 105, 51%)

(m/z = 164, 55%)

CH2H3CO

OCH3

CH3

CO_

CO_

(m/z = 77, 23%)

_.

O

O

H3CO

OCH3

CH3

m/z = 221, 19%

H3CO

OCH3

CH3O

m/z = 193, 13%

_OCH3

.

O

OOCH3

CH3

(m/z = 267, 39%)

+.

++

+

+

+

+

+

O

-CO

O

H3CO

OCH3

CH3 O

_

.

(18f)

Fig. 3.12 Mass fragmentation pattern of the compound (18f)

3.4 Synthesis of 6,8-dihydroxy-7-methyl-3-alkyl/aryl-3,4-dihydroisocoumarins

(19a-j)

Demethylation of the compounds (18a-j) was achieved using ethanethiol

(C2H5SH) and anhydrous aluminum chloride in good yield to furnish the corresponding

6,8-dihydroxy-7-methyl-3-alkyl/aryl-3,4-dihydroisocoumarins (19a-j). The 6,8-

dihydroxy-3,4-dihydroisocoumarins were characterized by the absence of both methoxy

singlets and presence of singlets for hydroxyl protons in 1H NMR spectrum. In

13C NMR

spectrum, the signals for methoxy carbons are also absent. The IR spectrum showed the

(O-H) absorption bands at 3445-3481 cm-1

. The physical constants and the IR spectral

data of the 6,8-dihydroxy-3,4-dihydroisocoumarins (19a-j) are shown in Table 3.22 and

the elemental analysis data in Table 3.23.

83

Scheme 3.5 Synthesis of 6,8-dihydroxy-7-methyl-3-alkyl/aryl-3,4-

dihydroisocoumarins (19a-j)

Table 3.22 Physical constants and FTIR spectral data of the compounds (19a-j)

Compds. M. P.

(°C) Rf

Yield

(%) υmax (cm

-1)

Ar-C-H Sp3 C-H C=O C=C O-H

19a 93-95 0.4 71 3019 2914 1723 1572 3474

19b 101-103 0.45 73 3021 2931 1719 1559 3456

19c 115-117 0.5 79 3017 2914 1727 1569 3467

19d 73-75 0.4 73 3010 2933 1717 1571 3445

19e 89-90 0.3 81 3009 2924 1720 1554 3481

19f 125-126 0.4 86 3020 2935 1722 1571 3477

19g 129-131 0.45 84 3023 2926 1720 1561 3488

19h 161-162 0.4 83 3018 2919 1724 1567 3467

19i 153-154 0.35 85 3017 2927 1719 1573 3479

19j 145-147 0.4 82 3010 2915 1716 1587 3459

Pet. Ether: Ethyl Acetate (4:1)

R=

18a = 18b = 18c =

18d = -CH2Cl 18e = -CH2OH

18f = 18g = 18h = 18i = 18j =Cl

OCH3

OCH3

OCH3

OCH3

OCH3

H3CO

CH2 CH3_

CH2 CH3_

CH2 CH3_

O

OOCH3

H3CO

CH3

R

H

(18a-j)

O

OOH

OH

CH3

RH

(19a-j)

C2H

5SH / 0-5 °C, 1h

AlCl3(Anhyd)

R is same as in (18a-j)

84

Table 3.23 Elemental analysis data of the compounds (19a-j)

Compounds

Molecular Formula

Calculated Found

C H C H

C13H16O4 (19a) 68.18 7.57 68.09 7.49

C15H20O4 (19b) 69.86 8.21 69.79 8.12

C17H24O4 (19c) 71.25 8.75 71.14 8.66

C11H11O4Cl (19d) 57.67 5.54 57.59 5.49

C11H12O5 (19e) 61.90 6.34 61.81 6.26

C16H14O4 (19f) 72.48 6.04 72.39 5.96

C16H13O4Cl (19g) 64.96 5.11 64.89 5.02

C17H16O5 (19h) 69.51 6.09 69.43 5.99

C18H8O6 (19i) 67.03 6.14 66.94 6.06

C19H20O7 (19j) 64.94 6.18 64.87 6.09

Table 3.24 1H and

13C NMR data of compound (19a)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 167.7

C-3 5.0, m 81.6

C-4 3.0, dd, J= 15.8, 3.7

3.5, dd, J=15.9, 12.1 44.3

C-4a ----- 143.1

C-5 6.9, s 129.2

C-6 ----- 149.4

C-7 ----- 118.3

C-8 ----- 149.4

C-8a ----- 138.8

C-1’ 2.4, m 37.3

C-2’ 1.2, m 18.1

C-3’ 0.9, t, J=7.5 10.2

6-OH 4.6, s -----

8-OH 4.6, s -----

7-CH3 2.7, s 29.4

The formation of 6,8-dihydroxy-7-methyl-3-propyl-3,4-dihydroisocoumarin (19a)

was confirmed by the presence of singlet at δ 4.6 ppm in 1H NMR spectrum for 6 and 8-

85

hydroxy hydrogens and disappearance of signals for methoxy hydrogens. It was also

supported in 13

C NMR spectrum by the presence of a peak at δ 167.7 ppm for lactonic

carbonyl carbon. The detailed 1H and

13C NMR data of the compound (19a) are presented

in Table 3.24.

The molecular ion peak which appeared at m/z 236 with 41% abundance,

confirmed the formation of dihydroisocoumarin (19a). By the removal of a molecule of

butanal from molecular ion, base peak at m/z 164 appeared. The fragmentation pattern of

the dihydroisocoumarin (19a) is shown in Fig. 13.

The detailed 1H and

13C NMR data of the dihydroisocoumarins (19e) and (19f)

are presented in Tables 3.25 and 3.26, respectively. The mass fragmentation pattern of

the (19e) and (19f) is shown in Fig. 14 and 15, respectively.

(m/z = 236, 41%)(m/z = 164, 100%)

O

OH

OH

CH3

CH2

_

(m/z = 72, 39%)

(m/z = 136, 47%)

OH

OH

CH3

CH2

CO_

CO_

(m/z =44, 62%)

.

O

OH

CH3

OH

_

_ .

O

O

OH

OH

CH3

(m/z = 193, 26%)

CO2_ OH

OH

CH3

(m/z = 149, 19%)

_CO

O

OH

CH3

CH3OH

(m/z = 208, 34%)

+.

++

+

+

+

+

+

O

O

OH

OH

CH3

CH3

OCH3

H

OCH3

H

CH3CH2CH3

CH3CH2CH2

.

CH3

(m/z = 115, 15%)

+

(19a)

.

.

.

-2OH.

Fig. 3.13 Mass fragmentation pattern of the compound (19a)

86

Table 3.25 1H and

13C NMR data of compound (19e)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 165.5

C-3 4.9, m 88.7

C-4 3.2, dd, J= 15.8, 3.5

3.6, dd, J=16.3, 12.1 39.1

C-4a ----- 140.5

C-5 6.7, s 121.5

C-6 ----- 149.2

C-7 ----- 128.9

C-8 ----- 149.2

C-8a ----- 138.6

C-1’ 5.4, dd, J=15.7, 12.1

4.8, dd, J=15.7, 3.8 65.3

O-H 2.1, s -----

6-OH 4.5, s -----

8-OH 4.5, s -----

7-CH3 2.9, s 30.2

(m/z = 224, 58%)(m/z = 164, 100%)

CH2

O

OH

OH

CH3

.

_

(m/z = 207, 28%)

(m/z = 136, 39%)

CH2OH

OH

CH3

CO_

CO_

(m/z =179, 31%)

.

_.

O

O

OH

OH

CH3

(m/z = 193, 45%)

CO2_ OH

OH

CH3

(m/z = 149, 23%)

_CO

O

OH

CH3

OHOH

(m/z = 196, 28%)

+.

++

+

++

+

+

O

O

OH

OH

CH3

OH

OOH

H

CH2OH

.

CH3

(m/z = 115, 13%)

+

(19e)

-OHO

O

OH

OH

CH3

O

OH

OH

CH3

.

.-2OH

Fig. 3.14 Mass fragmentation pattern of the compound (19e)

87

Table 3.26 1H and

13C NMR data of compound (19f)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 162.3

C-3 5.3, dd, J= 12.3, 3.8 81.5

C-4 3.0, dd, J= 12.0, 3.7

3.3, dd, J=15.7, 12.1 44.6

C-4a ----- 142.3

C-5 7.0, s 123.5

C-6 ----- 151.3

C-7 ----- 131.9

C-8 ----- 151.3

C-8a ----- 137.2

C-1’ ----- 138.1

C-2’ 7.9, d, J=5.8 129.4

C-3’ 7.7, dd, J=6.5, 5.8 126.3

C-4’ 7.5, dd, J=6.2, 5.6 119.5

C-5’ 7.7, dd, J=6.5, 5.8 126.3

C-6’ 7.9, d, J=5.8 129.4

6-OH 4.7, s -----

8-OH 4.7, s -----

7-CH3 2.5, s 28.1

O

O

OH

OH

CH3

(m/z = 270, 41%)(m/z = 164, 100%)

O

OH

OH

CH3

CH2

.

_

(m/z = 105, 51%)

(m/z = 136, 51%)

CH+

OH

OH

CH3

CO_

CO_

(m/z = 77, 23%)

_.

O

O

OH

OH

CH3

(m/z = 193, 21%)

OH

OH

CH3O

(m/z = 165, 15%)

.

O

OOH

CH3

(m/z = 253, 19%)

+.

++

+

+

+

+

+

O

-CO

O

OH

OH

CH3 O

_

.

-OH

(19f)

Fig. 3.15 Mass fragmentation pattern of the compound (19f)

88

4. BIOLOGIOCAL ACTIVITIES

A rapid advance in the development of new techniques for determining the

biological activity of synthetic and natural compounds has triggered a renaissance in the

drug development. Primary bioassay screening plays a very important role in the drug

development program. These screenings act as a tool to conduct activity directed

isolation of bioactive compounds for curing humans and animals. Primary screenings

provide first indication of bioactivities and thus help in the selection of lead compounds

for secondary screening for detailed pharmacological evaluation.

Isocoumarins, keto acids, 3,4-dihydroisocoumarins and 6,8-dihydroxy-3,4-

dihydroisocoumarins were tested for the following activities:

1. Antibacterial activity against ten different gram positive and gram

negative bacterial strains.

2. Antimalarial activity against Plasmodium falciparum

3. Cytotoxicity against human keratinocyte cell lines

4.1 Antibacterial Activity

Bacterial infections constitute one of the most serious situations in infectious

diseases. The detection and identification of these bacteria is one of the most important

functions of clinical microbiology. Isolation of an infectious agent from the patient with

disease is often not sufficient for determining proper therapy. Since the susceptibility of

many bacteria to antimicrobial agents cannot be predicted, testing individual pathogens,

against appropriate agent (with the most activity against the pathogen, the least toxicity to

the host, the least important on normal flora, appropriate pharmacologic characteristics

and most economical) can then be chosen allowing a more certain therapeutic outcome.

Antibacterial activity of the synthesized isocoumarins (16a-j), keto acids (17a-j),

3,4-dihydroisocoumarins (18a-j) and 6,8-dihydroxy-3,4-dihydroisocoumarins (19a-j) was

determined against various gram positive and gram negative bacterial strains by using

agar well diffusion. The purified samples were dissolved in DMSO 5mg/mL. DMSO is

the negative control and antibiotic chloramphenicol is the positive control in this in vitro

antibacterial study.

Ten bacterial strains Escherichia coli (E. c.), Klebsiella pneumonae (K. p.),

Lactobacillus bulgaricus (L. b.), Micrococcus luteus (M. l.), Pasteurella multocida (P.

89

m.), Proteus vulgaris (P. v.), Pseudomonas aeruginosa (P. a.), Salmonella typhi (S. t.),

Staphylococcus aureus (S. a.) and Staphylococcus epidermidis (S. e.) were selected for

this antibacterial assay. Micrococcus luteus, Staphylococcus aureus and Staphylococcus

epidermidis are the example of Gram positive and the remaining seven are Gram negative

bacteria. All of the tested microorganisms were maintained on nutrient agar at 4°C and

sub-cultured before use. The bacteria studied are clinically important ones causing

several infections and it is essential to overcome them through some active therapeutic

agents.

The antibacterial assay was performed by agar well diffusion method against

different bacterial strains142

. Each tested bacterium was sub-cultured in nutrient broth at

37°C for 24h. One hundred micro liters of each bacterium was spread with the help of

sterile spreader onto a sterile Muller-Hinton agar plate so as to achieve a confluent

growth. The plates were allowed to dry and wells (6mm diameter) were punched in the

agar with the help of cork borer. 0.1mL of each compound solution (5mg/mL) in DMSO

was introduced into the well and the plates were incubated overnight at 37°C.

The antimicrobial spectrum of the compounds was determined for the bacterial

species in terms of size of the zones around each well. The diameters of the zone of

inhibition produced by the compounds were compared with those produced by the

commercial antibiotic chloramphenicol (5mg/mL). This is the common antibiotic used

for the treatment of infections caused by gram positive and gram negative bacteria. The

control activity was deducted from the test and the results obtained were plotted. The

experiment was performed thrice to minimize the error and the mean values are

presented.

Antibacterial activity results of the isocoumarins (16a-j), keto acids (17a-j), 3,4-

dihydroisocoumarins (18a-j) and 6,8-dihydroxy3,4-dihydroisocoumarins are shown in

Tables 4.1, 4.2, 4.3 and 4.4, respectively.

90

Table 4.1 In vitro Antibcterial Activity of Isocoumarins (16a-j)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

16a 1 5 0 11 0 3 0 0 9 9

16b 1 8 0 12 0.5 0 2 0 8 11

16c 0 0 0 7 0 0 2 0 9 6

16d 1 2 1 8 0 1 0 0 8 1

16e 16 8 7 0.5 6 1 0 12 0.5 1

16f 3 3 4.5 2.5 5 1.5 2 0 2.5 2.5

16g 13 9 1 11 0 0 10 0 11 0

16h 1 0 11 2 10 12 0 13 0 11

16i 14 8 9 0 9 1 0 10 11 0

16j 1 0 1 10 0 0 11 0 0 11

Standard 18 10 13 13 12 13 13 14 13 13

Antibacterial activity results of the isocoumarins (16a-j) show that most of these

are more active against gram positive bacteria as compared to gram negative bacteria.

Some of them also possess activity against gram negative bacteria. Among the 3-alkyl

substituted isocoumarins, antibacterial activity of the 3-pentylisocoumarin is higher than

3-propylisocoumarin but less than 3-heptylisocoumarin. It reflects that antibacterial

activity increases by increasing the carbon chain length up to five carbons and then

decreases for seven carbons. Antibacterial activity does not directly correlate to

hydrophobicity.

The compound (16e) which possesses 3-hydroxymethyl substitution shows higher

activity against gram negative bacteria as compared to gram positive. Among the 3-

phenyl substituted isocoumarins, the most active are (16g) and (16h) which possess 3-

monomethoxyphenyl and dimethoxyphenyl substitution, respectively.

91

Table 4.2 In vitro Antibcterial Activity of Keto Acids (17a-j)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

17a 1 0 0 2 0 3 0 5 0 3

17b 1 2 0 1 0.5 0 2 0 1 3

17c 0 0 0 3 0 0 2 0 12 1

17d 1 2 1 0.5 0 1 0 0 2 1

17e 15 1 3 0.5 11 1 0 12 0.5 1

17f 3 3 4.5 2.5 1 1.5 2 0 2.5 2.5

17g 1 9 1 0.5 0 0 1 0 11 0

17h 1 0 11 2 10 12 0 13 0 1.5

17i 0.5 1 2 0 3 1 0 1 2 0

17j 1 0 1 0.5 11 0 1 0 0 1.5

Standard 18 10 13 13 12 13 13 14 13 13

Antibacterial activity results of the keto acids (17a-j) are presented in Table 4.2

which shows that most of the keto acids are inactive against the selected bacterial strains.

After hydrolysis of the lactonic ring, antibacterial activity is greatly suppressed. Presence

of carbonyl and carboxylic acid polar functionalities in keto acids (17a-j) decreases their

antibacterial activity. It may partly be due to the fact that the cell barriers of the

microorganisms are nonpolar in nature so highly polar compounds can not easily pass

these hydrophobic barriers.

92

Table 4.3 In vitro Antibcterial Activity of 3,4-Dihydroisocoumarins (18a-j)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

18a 1 7 0 11 0 3 0 0 10 12

18b 1 9 0 12 0.5 0 2 0 9 11

18c 0 0 0 9 0 0 2 0 5 4

18d 1 2 1 6 0 1 0 0 5 1

18e 16 9 11 0.5 9 1 0 13 0.5 1

18f 0 1 2.5 0.5 10 11.5 2 0 2.5 11

18g 15 9 1 11.5 0 11 0 0 12 0

18h 0.5 7 12 2 10 12 0 13 0 11

18i 14 8 9 0 9 1 0 10 11 0

18j 1 0 1 11 0 0 12 0 0 9

Standard 18 10 13 13 12 13 13 14 13 13

*Activity of each sample is measured by subtracting the activity of DMSO. Escherichia

coli, Klebsiella pneumonae, Lactobacillus bulgaricus, Micrococcus luteus, Pasteurella

multocida, Proteus vulgaris, Pseudomonas aeruginosa, Salmonella typhi, Staphylococcus

aureus and Staphylococcus epidermidis)

Antibacterial activity results of the 3,4-dihydroisocoumarins (18a-j) shows that

these are more active against gram positive bacteria as compared to gram negative

bacteria. Some of them also possess antibacterial activity against gram negative bacteria.

Among the 3-alkyl substituted isocoumarins antibacterial activity of the 3-

pentylisocoumarine is higher than 3-propylisocoumarine but less than 3-

heptylisocoumarine. It reflects that anti bacterial activity increases by increasing the

carbon chain length up to five carbon and then decreases for seven carbon. This is similar

behavior as possessed by isocoumarins (16a-j)

The compound (18e) possesses 3-hydroxymethyl substitution having higher

activity against gram negative bacteria as compared to gram positive. Among the 3-

phenyl substituted isocoumarins the most active are (18g) and (18h) which possesses 3-

monomethoxyphenyl and dimethoxyphenyl substitution respectively.

93

Table 4.4 In vitro Antibcterial activity of 6,8-Dihydroxy3,4-dihydroisocoumarins

(19a-j)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

19a 14 0.5 3 1 11 10 9 0 0 1

19b 12 3 5 1 10 12 10 13 0.5 1.5

19c 0 9 10 0.5 0 0 2 0 1 0.5

19d 1 2 1 1 0 1 10 0 5 1

19e 15.5 9 12 0.5 11 1 0 13 0.5 1

19f 0 1 2.5 0.5 10 11.5 2 0 2.5 12

19g 15 9 1 11.5 0 11 0 0 12 0

19h 0.5 7 12 2 10 12 0 13 0 11

19i 14 8 9 0 11 1 0 12 11 0

19j 1 0 1 11 0 10 12 0 0 11

Standard 18 10 13 13 12 13 13 14 13 13

*Activity of each sample is measured by subtracting the activity of DMSO. Escherichia

coli, Klebsiella pneumonae, Lactobacillus bulgaricus, Micrococcus luteus, Pasteurella

multocida, Proteus vulgaris, Pseudomonas aeruginosa, Salmonella typhi, Staphylococcus

aureus and Staphylococcus epidermidis)

The antibacterial activity results of the 6,8-dihydroxy-3,4-dihydroisocoumarins

(19a-j) show that most of these are more effective against gram negative bacteria as

compared to gram positive. Some of them also possess activity against gram positive

bacterial strains.

The comparison of the antibacterial activity among the four series of the

synthesized compounds indicates that isocoumarins (16a-j) and 3,4-dihydroisocoumarins

(18a-j) are more active against gram positive bacteria than gram negative. But the 6,8-

dihydroxy-3,4-dihydroisocoumarins are more active against gram negative than gram

positive. Among all these compounds the keto acids (17a-j) are the least active. Although

the 3-alkyl substituted isocoumarins and dihydroisocoumarins possess antibacterial

activity but the compounds which possess 3-substituted phenyl ring are more active than

the 3-alkyl substituted ones.

94

4.2 Antimalarial Activity

Malaria is the worldwide most important parasitic disease with an incidence of

almost 300 million clinical cases and over one million deaths per year. Malaria indirectly

contributes to illness and deaths from respiratory infections, diarrhoeal disease and

malnutrition. Plasmodium falciparum, the potentially lethal malarial parasite, has shown

itself capable of developing resistance to nearly all used antimalarial drugs and resistant

strains have rapid extension143

.

The lost of effectiveness of chemotherapy constitute the greatest threat to the

control of malaria. Therefore, to overcome malaria, new knowledge, products and tools

are urgently needed; especially new drugs are required144

. All the synthesized compounds

are evaluated for their potential against malarial parasite Plasmodium falciparum.

In vitro antimalarial assay was performed in duplicate in a 96 well microtiter

plate. The assay was based on assessing the inhibition of schizont maturation.

For the cultivation of Plasmodium falciparum, RPMI 1640 supplemented with 25 mM

HEPES, 1% D-glucose, 0.23% sodium bicarbonate and 10% heat inactivated human

serum.

A stock solution of 5mg/ml of each of the test samples was prepared in DMSO

and subsequent dilutions were prepared with culture medium. The final concentrations

ranged from 1.5 µg/mL to 100 µg/mL. The diluted samples in 20µL volume were added

to the test well and the culture plates were incubated at 37 °C for 36 to 40 hrs. After

incubation, contents of the wells were harvested and stained for 30min in a 2% Giemsa

solution pH 7.2, after that the developed schizonts were counted.

The antimalarial activity of the synthesized compounds was expressed by the

inhibitory concentrations 50 (IC50), representing the concentration of the drug that

induced a 50% parasitaemia decrease and 100% schizonts maturation inhibition.

Chloroquine phosphate is the positive control with IC50 value 0.2 µg/mL and 12.5 µg/mL

values for 100% schizont maturation inhibition.

95

Table 4.5 Antimalarial Activity of isocoumarins (16a-j)

Compds. IC50 µg/ml 100%SMI (µg/ml)

16a 5 15

16b 2 9.5

16c 16 89

16d 65 -

16e 53 -

16f 34 -

16g 25 -

16h 4 19

16i 62 -

16j 36 -

Standard 0.2 12.5

SMI, Schizont Maturation Inhibition; IC50, Inhibition Concentration 50

The results of the in vitro antimalarial activity of the isocoumarins (16a-j) are

presented in table 4.5. The results showed that isocoumarins possess moderate to potent

antimalarial activity. The 3-alkyl substituted isocoumarins are active against the tested

malarial strain. The antimalarial activity of these compounds depends upon the size of the

3-alkyl group. Activity increases from 3-propylisocoumarin (16a) to 3-pentylisocoumrin

(16b) but decreases to 3-heptylisocoumarin (16c). The antimalarial activity of these

compounds can be correlated to their hydrophobicity, the activity increases with the

increase of the hydrophobic carbon chain but hydrophobicity must be in certain limits.

The most interesting antiplasmodial activity is obtained by isocoumarins (16b) (IC50 2

µg/ml). It exhibit complete inhibition of the schizonts maturation at 9.5µg/ml. This

compound is most potent than all other tested isocoumarins (6a-j).

Among the 3-phenyl substituted isocoumarins the most active is (16h) which

possess 4-methoxyphenyl substitution at 3 positions with IC50 4 µg/ml. The results

showed complete inhibition of the schizonts maturation at 19 µg/ml for this

isocoumarins.

96

Table 4.6 Antimalarial Activity of Keto Acids (17a-j)

Compd IC50 µg/ml 100%SMI (µg/ml)

17a 45 -

17b 32 -

17c 56 -

17d 78 -

17e 67 -

17f 77 -

17g 65 -

17h 19 -

17i 69 -

17j 53 -

Standard 0.2 12.5

SMI, Schizont Maturation Inhibition; IC50, Inhibition Concentration 50

Table 4.6 showed the in vitro antimalarial activity results of the keto acids (17a-

j). Most of these keto acids are inactive against the selected malarial strain. The

hydrolysis of lactonic ring introduced polar carboxylic acid and keto group in the keto

acids (17a-j). These polar functional groups decrease the antiplasmodial activity of these

keto acids. Presence of the polar functionalities of the keto acids decreases their lipophilic

character decreasing the penetration of these compounds through cell barrier.

Isocoumarins (16a-j) have high antimalarial activity as compared to keto acids

(17a-j). In conclusion, the antimalarial activity is highly suppressed after the hydrolysis

of the lactonic ring of isocoumarins to keto acids. The presence of the polar keto group

and carboxylic acid group results in the loss of antimalarial activity.

97

Table 4.7 Antimalarial Activity of 3,4-Dihydroisocoumarins (18a-j)

Compd IC50 µg/ml 100%SMI (µg/ml)

18a 3.5 13.5

18b 1.2 7.5

18c 18 78

18d 57 -

18e 48 -

18f 31 -

18g 22 -

18h 2.6 15.6

18i 58 -

18j 42 -

Standard 0.2 12.5

SMI, Schizont Maturation Inhibition; IC50, Inhibition Concentration 50

The results of the in vitro antimalarial activity of the 3,4-dihydroisocoumarins

(18a-j) are presented in Table 4.7. The results showed that 3,4-dihydroisocoumarins are

more active as compared to isocoumarins (16a-j). The 3-alkyl substituted 3,4-

dihydroisocoumarins are active against the tested malarial strain. The antimalarial

activity of these compounds depends upon the size of the 3-alkyl group. Activity

increases from 3-propyl-3,4-dihydrolisocoumarin (18a) to 3-pentyl-3,4-

dihydroisocoumarin (18b) but decreases to 3-heptyl-3,4-dihydroisocoumarin (18c). The

antimalarial activity of these compounds can be related to their hydrophobicity, activity

increases with the increase of the hydrophobic carbon chain but hydrophobicity must be

in certain limits. The most interesting antiplasmodial activity is obtained for isocoumarin

(18b) (IC50 1.2 µg/ml). It exhibits complete inhibition of the schizonts maturation at

7.5µg/mL. This compound is most potent of all other tested dihydroisocoumarins (18a-j).

Among the 3-substituted phenyl 3,4-dihydroisocoumarins, the most active is

(18h) which possesses 4-methoxyphenyl substitution at 3 position with IC50 2.6 µg/mL.

The results showed complete inhibition of the schizonts maturation at 15.6 µg/mL for this

3,4-Dihydroisocoumarins.

98

Table 4.8 Antimalarial Activity of 6,8-Dihydroxy-3,4-dihydroisocoumarins (19a-j)

Compd IC50 µg/ml 100%SMI (µg/ml)

19a 1.3 8.4

19b 1.5 7.2

19c 21 78

19d 53 -

19e 44 -

19f 37 -

19g 27 -

19h 2.1 12.4

19i 43 -

19j 39 -

Standard 0.2 12.5

SMI, Schizont Maturation Inhibition; IC50, Inhibition Concentration 50

The results of the in vitro antimalarial activity of the 6,8-Dihydroxy-3,4-

dihydroisocoumarins (19a-j) are presented in table 4.8. The results showed that 6,8-

Dihydroxy-3,4-dihydroisocoumarins (19a-j) are more active as compared to

isocoumarins (16a-j), keto acids (17a-j) and 3,4-Dihydroisocoumarins (18a-j). The 3-

alkyl substituted derivatives showed moderate to potent activity against the tested

malarial strain. The antimalarial activity of these compounds depends upon the size of the

3-alkyl group. Activity decreases from 3-propy-6,8-dihydroxy-3,4-dihydrolisocoumarin

(19a) to 3-pentyl-6,8-dihydroxy-3,4-dihydroisocoumrin (19b) and 3-heptyl-6,8-

dihydroxy-3,4-dihydroisocoumarin (19c). The antimalarial activity of these compounds

can be related to carbon chain length, activity decreases by increasing the carbon chain

length of three alkyl group. The most interesting antiplasmodial activity is obtained by

isocoumarins (19a) (IC50 1.3 µg/ml). It exhibit complete inhibition of the schizonts

maturation at 8.4µg/ml. This compound is most potent than all other tested 3-alkyl-6,8-

dihydroxy-3,4-dihydroisocoumarins (19a-j).

Among the 3-phenyl substituted 3,4-Dihydroisocoumarins the most active is

(19h) which possess 4-methoxyphenyl substitution at 3 positions with IC50 2.1 µg/ml.

99

The results showed complete inhibition of the schizonts maturation at 12.4 µg/ml for this

analogue.

• Keto acids are inactive against tested strains of malarial parasite.

• 6,8-Dihydroxy-3,4-dihydroisocoumarins (19a-j) are most effective as compared

to isocoumarins (16a-j) and 3,4-dihydroisocoumarins (18a-j).

• 6,8-Dihydroxy-3,4-dihydroisocoumarins having 3-alkyl substitution are more

effective as compared to 3-aryl substituted derivatives.

• The size of carbon chain length at position 3 also plays important role in

antimalarial activity of these compounds. Activity increases when n-propyl and n-

pentyl group are present.

100

4.3 Cytotoxicity

The neutralrottest was in line with the protocol of the National Institute of Health

(NIH) implemented. It is of vital dye Neutral from living cells and then protonated. The

cell acts as an ion trap, whereby the dye is no longer out of can diffuse. By destroying the

cells, the neutral released and can be determined photometrically. It represents the

absorption is a measure of the vitality of the cells. The lower the absorption, the fewer

living cells were present.

For testing immortalize human keratinocyte cell line (HaCaT) was used. The

incubation with the test substances was made over three days and the experiments were

carried out in two independent experiments performed with several parallels.

The stock solutions of the substances in DMSO were prepared. The tested

concentration range was between 1.56 to 100 µM. the concentration of the solvent

DMSO was tested at all concentrations at 0.1%. Etoposide was used as a positive control

in a concentration of 10 µM with IC50 0.8 µM and DMSO as a negative control.

4.3.1 Cytotoxic Activity of the isocoumarins (16a-j)

The Cytotoxic activity results of the isocoumarins (16a-e) and (16f-j) are

presented in figures 4.1 and 4.2 respectively. The activity of all these compounds

increases as the concentration of the compounds increases and they show the maximum

activity at highest concentration. Among the 3-alkyl substituted isocoumarins the most

active is the isocoumarins (16b) having 3-pentyl substitution.

The isocoumarin (16e) possess 3-hydroxymethyl group is the most active which

show only the 20% viability of the infected cells at concentration of 100 μM. The 3-

hydroxymethyl group plays vital role in the activity of this compound and may be

important for the receptor binding. This derivative has the structural similarities with the

well known antitumor agent cytogenin.

101

Fig. 4.1 Cytotoxic activity results of the isocoumarins (16a-e)

The isocoumarins (16f-j) show moderate to potent cytotoxic activity against the

immortalized human keratinocyte cell lines. When we compare the cytotoxicity of the 3-

phenyl substituted isocoumarins (16f-j) the isocoumarins (16h) and (16i) have higher

activity than the others. These two derivatives possess monomethoxy and dimethoxy

phenyl ring at 3-position.

The compound (16h) having 4-methoxyphenyl substitution at 3-position show a

40% viability of the infected cells at a concentration of 50μM and 20% viability at

highest concentration 100μM. The isocoumarin (16i) show a 50% decrease of the

viability of the infected cells at highest tested concentration 100 μM and is less active

than the isocoumarin (16h).

Fig. 4.2 Cytotoxic activity results of the isocoumarins (16f-j)

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bilit

y [

%]

Concentration [uM]

16a

16b

16c

16d

16e

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bil

ity [

%]

Concentration [uM]

16f

16g

16h

16i

16j

102

4.3.2 Cytotoxic Activity of the Keto Acids (17a-j)

The cytotoxicity of the keto acids (17a-e) and (17f-j) are summarized in figures 3

and 4 respectively. From the graphs shown in figures 4.3 and 4.4 it is clear that the keto

acids show only 20-30% decrease of the viability of the infected cells. The results

indicates that the keto acids (17a-j) are inactive against the immortalized human

keratinocyte cell lines (HaCaT). The presence of the carboxylic acid and keto

functionalities decreases their penetration through the plasma membrane due to which

they show no activity against the immortalized human keratinocyte cell lines.

Fig. 4.3 Cytotoxic activity results of the keto Acids (17f-j)

Fig. 4.4 Cytotoxic activity results of the Keto Acids (17f-j)

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bilit

y [

%]

Concentration [uM]

17a

17b

17c

17d

17e

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bilit

y [

%]

Concentration [uM]

17f

17g

17h

17i

17j

103

4.3.3 Cytotoxic Activity of the 3,4-Dihydroisocoumarins (18a-j)

The Cytotoxic activity results of the 3,4-Dihydroisocoumarins (18a-e) and (18f-j)

are presented in figures 4.5 and 4.6 respectively. They show moderate to potent

cytotoxicity against the infected cell lines. The activity of all these compounds increases

as the concentration of the compounds increases and they show the maximum activity at

highest concentration. Among the 3-alkyl substituted isocoumarins the most active is the

3-pentyl-3,4-dihydroisocoumarin (18b) which show 205 viability of the infected cells at

100 μM.

The 3,4-Dihydroisocoumarin (18e) possess 3-hydroxymethyl group is the most

active which show less than 20% viability of the infected cells at concentration of 100

μM. The reduction of the double bond between C-3, C-4 and 3-hydroxymethyl group

plays vital role in the activity of this compound and may be important for the receptor

binding.

Fig. 4.5 Cytotoxic activity results of the 3,4-Dihydroisocoumarins (18a-e)

Fig. 4.6 Cytotoxic activity results of the 3,4-Dihydroisocoumarins (18f-j)

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bil

ity [

%]

Concentration [uM]

18a

18b

18c

18d

18e

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bil

ity [

%]

Concentration [uM]

18f

18g

18h

18i

18j

104

Among the 3,4-Dihydroisocoumarins (18f-j) the compound (18h) and (18i) have

higher activity than the others. These two derivatives possess monomethoxy and

dimethoxy phenyl ring at 3-position.

The compound (18h) having 4-methoxyphenyl substitution at 3-position show a

50% decrease of the viability of the infected cells at a concentration of 27 μM and less

than 20% viability at highest concentration 100 μM. The 3,4-dihydroisocoumarin (18i)

show a 50% decrease of the viability of the infected cells at highest tested concentration

100 μM and is less active than the isocoumarin (18h).

4.3.4 Cytotoxic Activity of the 6,8-dihydroxy-3,4-Dihydroisocoumarins (19a-j)

The figure 4.7 presented the cytotoxicity of the 6,8-Dihydroxy-3,4-

dihydroisocoumarins (19a-e). The compounds (19a), (19b) and (19e) show higher

activity. Among the 3-alkyl substituted compounds the activity decreases as size of the

alkyl group increases. The compounds (19a) which possess 3-propyl substituent show a

50 % decrease of the viability of the infected cells at a concentration 10 μM. The other

two which possess 3-pentyl and 3-heptyl substituents show a 50% decrease at higher

concentrations. Activity of these derivatives can be related to their hydrophobicity,

greater the hydrophobicity lesser will be the cytotoxic activity. The compound (19e)

which possesses 3-hydroxymethyl group show a 50% decrease of the viability of the

infected cells at 15 μM and less than 10% viability at highest tested concentration.

Fig. 4.7 Cytotoxic activity results of the 6,8-Dihydroxy-3,4-Dihydroisocoumarins

(19a-e)

0

20

40

60

80

100

10 25 50 100 1000

Via

bil

ity [

%]

Concentration [uM]

19a

19b

19c

19d

19e

105

The cytotoxic activity results of the 3-phenyl substituted derivatives (19f-j) are

presented in figure 4.8. Among these the compound (19h) and (19i) show higher activity.

The compound (19i) which possesses the dimethoxy substituted phenyl ring show a 50%

decrease of the viability of the infected cells at a concentration 50 μM and about 30%

infected cell are viable at highest tested concentrations. The compound (19h) which

possesses monomethoxy substituted phenyl ring shows the comparable results as the

compound (19i).

In a comparison of the 3-alkyl substituted 6,8-dihydroxy-3,4-dihydroisocoumarins

and 3-phenyl substituted the 3-alkyl substituted derivatives has higher activity. It may be

due to the fact that the 3-alkyl group is important in the absorption of these derivatives

through the cell memberane.

Fig. 4.8 Cytotoxic activity results of the 6,8-dihydroxy-3,4-dihydroisocoumarins

(19f-j)

Following are some general conclusions derived from the results of the cytotoxic

activity of the synthesized compounds.

Keto acids having no cytotoxic activity against human keratinocytes cell lines

(HaCaT).

Among 6,8-dihydroxy-3,4-dihydroisocoumarins (19a-j), 6,8-dimethoxyisocoumarins

(16a-j) and 6,8-Dimethoxy-3,4-dihydroisocoumarins (18a-j), compounds (19a-j) have

higher cytotoxic activity.

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bilit

y [

%]

Concentration [uM]

19f

19g

19h

19i

19j

106

• 3-alkyl substituted analogous are more active as compared to 3-phenyl

substituted. Compound having 3-(4’-methoxyphenyl) substitution have high

potency.

• Among 6,8-Dihydroxy-3,4-dihydroisocoumarins, the derivative possess 3-

hydroxymethyl group is more potent as compared to all other members, as this

member is the structural analogue of a well known cytotoxic agent cytogenin.

107

5.1 Synthesis of Stellatin

The solvents were purified and dried according to the standard procedures before

using. The dried solvents were stored under molecular sieves (4 Å). Standard procedures

were employed for the purification and drying of solvents. Melting points were recorded

using a digital Gallenkamp (SANYO) model MPD BM 3.5 apparatus and are

uncorrected. FTIR spectra were recorded using an FTS 3000 MX spectrophotometer, 1H

NMR and 13

C NMR spectra were determined as CDCl3 solutions at 300 MHz on a Bruker

AM-300 spectrophotometer, mass spectra (EI, 70eV) on a GCMS instrument, and

elemental analyses with a LECO-183 CHNS analyzer. All the compounds were purified

by thin layer chromatography using silica gel HF-254 from Merck.

5.1.1 Methyl (3, 5-dimethoxy-4-methylphenyl) acetate (1)

A stirred solution of (3,5-dimethoxy-4-methyl phenyl) acetic acid (5.0g, 23.8

mmol) in dry methanol (30 mL) was treated drop wise with conc. H2SO4 (5mL). The

mixture was refluxed for 8-9h. The reaction was monitored by TLC. After the completion

of reaction, mixture was concentrated to 55mL and extracted with ethyl acetate (3x50

mL). The extract was washed with saturated brine, dried and concentrated to give crude

oil which was distilled to afford Methyl (3,5-dimethoxy-4-methyl phenyl) acetate (1)

(4.7g, 88.18%); Rf: 0.7 (petroleum ether and ethyl acetate, 4:1); m. p. 38-40 °C, IR

(KBr): 3023 (C-H), 1734 (C=O), 1573 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm): 7.45 (2H,

s, H-2, H-6), 3.96 (6H, s, 3-OCH3, 5-OCH3), 3.54 (2H, s, Ar-CH2), 3.47 (3H, s,

COOCH3), 2.55 (3H, s, 4-CH3); 13

C NMR (CDCl3 δ ppm): 168.23 (C=O), 132.54 (C3,

C5), 128.37 (C2, C6), 119.42 (C4), 112.21 (C1), 68.55 (Ester OCH3), 55.34 (Ar-OCH3),

36.91 (CH2), 28.63 (Ar-CH3); MS (70eV): m/z (%); 224 [M]+.

(46), 193 (43), 165 (100),

59 (12); Anal. calcd for C12H16O4: C, 64.28 H, 7.14 O, 28.57 Found: C, 64.02 H, 6.96

O, 28.35.

5.1.2 Methyl (2-formyl-3,5-dimethoxy-4-methylphenyl) acetate (2)

Phosphorus oxychloride (1.61g, 10.0mmol) was added dropwise in to a stirred

solution of methyl (3,5-dimethoxy-4-methyl phenyl) acetate (1) (2.0g, 8.9mmol) in

freshly distilled DMF (10mL) at 55 °C. Reaction mixture was heated at about 100 °C for

2 hr and stirred overnight at room temperature. Then poured the reaction mixture into

aqueous solution of sodium acetate (10%, 10mL) and shake vigorously. Methyl (2-

108

formyl-3,5-dimethoxy-4-methyl phenyl) acetate (2) was precipitated out as yellowish

precipitates (1.9g, 84%); Rf: 0.55 (petroleum ether and ethyl acetate, 4:1); m. p. 51-53

°C; IR (KBr): 3029 (C-H), 1722 (C=O), 1690 (CHO), 1545 (C=C) cm-1

; 1H NMR,

(CDCl3, δ ppm ): 9.75 (1H, s, CHO), 7.96 (1H, s, H-6), 3.42 (3H, s, 3-OCH3), 3.25 (3H,

s, 5-OCH3), 3.11 (3H, s, CO2CH3), 2.92 (2H, s, Ar-CH2), 2.80 (3H, s, 4-CH3); 13

C NMR

(CDCl3, δ ppm ): 179.32 (Aldehyde C=O), 162.43 (Ester C=O), 136.76 (C3, C5), 131.89

(C2), 126.21 (C6), 121.33 (C4), 117.54 (C1), 61.63 (Ester OCH3), 57.34 (Ar-OCH3),

39.12 (Ar-CH2), 32.08 (4-CH3); MS (70eV): m/z (%); 252 [M]+ (25), 251 (65), 224 (49),

223 (34), 165 (100), 29 (31); Anal. calcd. for C13H16O5: C, 61.90 H, 6.34 O, 31.74

Found: C, 61.67 H, 6.16 O, 31.56.

5.1.3 2,4-Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid (3)

Methyl (2-formyl-3, 5-dimethoxy-4-methyl phenyl) acetate (2) (6.3g, 25.0 mmol)

and sulfamic acid (8.3g, 86.0 mmol) in 150ml H2O:THF:DMSO (20:1:1) at 0°C was

treated with NaClO2 (7.24g, 80.0 mmol) in 20mL H2O. The reaction mixture was stirred

for 20min at 0°C and then diluted with ethyl acetate (100mL), washed with saturated

aqueous ammonium chloride (2 x 130mL) saturated aqueous sodium chloride (130mL).

Organic layer was dried over anhydrous sodium sulfate and evaporated to afford 2,4-

Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid (3) (6.6g, 79%); Rf: 0.4

(petroleum ether and ethyl acetate, 4:1); m. p. 164-166 °C; IR (KBr): 3265 (O-H), 3037

(C-H), 1734 (C=O), 1715 (COOH), 1562 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm ): 8.19

(1H, s, COOH), 7.66 (1H, s, H-6), 3.82 (3H, s, 3-OCH3), 3.67 (3H, s, 5-OCH3), 3.63 (3H,

s, CO2CH3), 2.54 (2H, s, Ar-CH2), 2.25 (3H, s, 3-CH3); 13

C NMR (CDCl3, δ ppm ):

197.78 (Carboxylic C=O), 168.56 (Ester C=O), 139.32 (C3, C5), 134.37 (C2), 127.13

(C6), 120.62 (C4), 114.17 (C1), 66.09 (Ester OCH3), 55.41 (Ar-OCH3), 35.04 (Ar-CH2),

29.88 (3-CH3); MS (70eV): m/z (%); 268 [M]+ (32), 251 (51), 224 (65), 165 (100), 45

(25); Anal. calcd for C13H16O6: C, 58.20 H, 5.97 O, 35.82 Found: C, 58.04 H, 5.76 O,

35.59.

5.1.4 2,4-Dimethoxy-6-(2-hydroxyethyl)-3-methylbenzoic acid (4)

2,4-Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-methylbenzoic acid (3) (0.5g, 1.86

mmol) and sodium borohydride (0.84g, 22.32 mmol) were suspended in freshly distilled

THF (10mL). The reaction mixture was stirred for 15min at 65 °C and then added

109

methanol (10 mL) dropwise during 30min. The mixture was refluxed for 4 h then cooled

to room temperature and treated with saturated ammonium chloride solution (10 mL).

Stirring was continued for 1 h then acidified with dilute hydrochloric acid and extracted

with ethyl acetate (3 x 20mL). The extract was dried over anhydrous sodium sulfate and

evaporated to afford 2,4-dimethoxy-6-(2-hydroxyethyl)-3-methylbenzoic acid (4) (0.35g,

78%); Rf: 0.3 (petroleum ether and ethyl acetate, 4:1); m. p. 72-74 °C, IR (KBr): 3481

(O-H), 3009 (C-H), 1710 (C=O), 1574 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm ): 8.22 (1H,

s, COOH), 7.48 (1H, s, H-5), 4.21 (2H, t, J=3.8, H-1’), 3.90 (3H, s, 2-OCH3), 3.75 (3H,

s, 4-OCH3), 2.65 (2H, t, J=3.8Hz, H-2’ ) 2.51 (3H, s, 3-CH3); 13

C NMR (CDCl3, δ ppm):

190.5 (COOH), 166.2 (C2), 162.4 (C4), 141.3 (C6), 108.1 (C3), 105.6 (C1), 101.2 (C5),

63.2 (C2’), 56.4 (2-OCH3,4-OCH3), 32.3 (C1’), 28.8 (3-CH3); MS (70eV): m/z (%); 240

[M]+ (32), 223 (24), 196 (43), 165 (100), 45 (19), 31 (37); Anal. Calcd. for C12H16O5: C,

60.00 H, 6.66 Found: C, 59.87 H, 6.57.

5.1.5 6,8-Dimethoxy-7-methyl-3,4-dihydro-1H-isochromen-1-one (5)

2,4-dimethoxy-6-(2-hydroxyethyl)-3-methylbenzoic acid (4) (1.0g, 4.16 mmol)

was dissolved in acetic anhydride (5mL) and refluxed for 1 h. Then reaction mixture was

poured into ice cold water and extracted with ethyl acetate (3 x 20mL). The combined

ethyl acetate extract was washed with (1% NaHCO3) and then with water. The ethyl

acetate was evaporated under reduced pressure to afford 6,8-dimethoxy-7-methyl-3,4-

dihydro-1H-isochromen-1-one (5). Yield 78% Rf: 0.6 (petroleum ether and ethyl acetate,

4:1); m. p. 145-147°C; IR (KBr): 3010 (C-H), 1702 (C=O), 1591 (C=C) cm-1

; 1H NMR

(CDCl3, δ ppm ): 7.48 (1H, s, H-5), 4.25 (2H, t, J=3.6, H-3), 3.90 (6H, s, 6-OCH3, 8-

OCH3), 2.66 (3H, s, 7-CH3), 2.56 (2H, t, J=3Hz, H-4); 13

C NMR (CDCl3, δ ppm ) 163.9

(C1), 152.3 (C6, C8), 140.8 (C4a), 134.6 (C8a), 108.9 ( C7), 103.7 (C5), 65.9 (C3), 56.4

(6-OCH3, 8-OCH3), 27.4 (C4), 26.2 (7-CH3); MS (70eV): m/z (%); 222 [M]+ (52), 194

(45), 192 (100), 164 (31), 30 (21); Anal. Calcd. for C12H14O4 C, 64.86 H, 6.30 Found C,

64.75 H, 6.19 O.

5.1.6 6,8-Dimethoxy-7-(bromomethyl)-3,4-dihydro-1H-isochromen-1-one (6)

To a stirred solution of 6,8-dimethoxy-7-methyl-3,4-dihydro-1H-isochromen-1-

one (5) (0.5g, 2.25 mmol) in dry carbon tetrachloride (10mL) was added N-

bromosuccinimide (0.6g, 3.37 mmol) and benzoyl peroxide (7.5mg). The reaction

110

mixture was refluxed for 6 h, then cooled, filtered and washed with a little carbon

tetrachloride. Solvent was rotary evaporated to get 6,8-dimethoxy-7-(bromomethyl)-3,4-

dihydro-1H-isochromen-1-one (6). (Yield 81%); Rf: 0.6 (petroleum ether and ethyl

acetate, 4:1); m. p. 91-93°C; IR (KBr): 3013 (C-H), 2926 (Ar-C-H), 1718 (C=O), 1582

(C=C) cm-1

; 1H NMR (CDCl3, δ ppm ): 7.50 (1H, s, H-5), 4.85 (2H, s, CH2-Br), 4.24 (2H,

t, J=3.6, H-3), 3.90 (6H, s, 6-OCH3, 8-OCH3), 2.58 (2H, t, J=3.6, H-4); 13

CNMR

(CDCl3, δ ppm ): 169.2 (C1), 155.4 (C6, C8), 144.1 (C4a), 110.7 ( C7), 106.2 (C8a),

104.6 (C5), 67.4 (C3), 53.7 (6-OCH3, 8-OCH3), 39.5 (CH2-Br), 26.4 (C4). MS (70eV):

m/z (%); 200 [M]+ (24), 302 [M+2] (24), 272 (39), 221 (19), 191 (100), 30 (28); Anal.

calcd. for C12H13O4Br C, 47.84 H, 4.31 Found C, 47.72 H, 4.23.

5.1.7 7-(Hydroxymethyl)-6,8-dimethoxy-3,4-dihydro-1H-isochromen-1-one (7)

6,8-Dimethoxy-7-(bromomethyl)-3,4-dihydro-1H-isochromen-1-one (6) (1.0g,

3.32 mmol) was dissolved in a mixture of water and acetone (10 mL, 1:1). The reaction

mixture was refluxed for 1 h then most of the acetone was rotary evaporated. Then

poured into ice cold water and the solid was filtered, washed with water and dried to

afford 7-(hydroxymethyl)-6,8-dimethoxy-3,4-dihydro-1H-isochromen-1-one (7), (Yield

77%); Rf: 0.4 (petroleum ether and ethyl acetate, 4:1); m. p. 136-138°C; IR (KBr): 3467

(O-H), 3010 (C-H), 2919 (Ar-C-H), 1714 (C=O), 1587 (C=C) cm-1

; 1H NMR (CDCl3, δ

ppm ): 7.62 (1H, s, H-5), 4.88 (2H, s, CH2-OH), 4.20 (2H, t, J=3.7, H-3), 3.90 (6H, s, 6-

OCH3, 8-OCH3), 2.56 (2H, t, J=3.7, H-4), 2.36 (1H, s, O-H); 13

C NMR (CDCl3, δ ppm ):

168.6 (C1), 157.3 (C6, C8), 142.1 (C4a), 115.2 ( C7), 105.2 (C8a), 102.6 (C5), 64.5 (C3),

55.7 (6-OCH3, 8-OCH3), 48.6 (CH2-OH), 27.4 (C4); MS (70eV): m/z (%); 238 [M]+ (49),

221 (31), 210 (55), 208 (100), 30 (19); Anal. calcd. for C12H14O5 C, 60.50 H, 5.88 Found

C, 60.38 H, 5.76.

5.1.8 8-Hydroxy-7-(hydroxymethyl)-6-methoxy-3,4-dihydro-1H-isochromen-1-one (8)

7-(Hydroxymethyl)-6,8-dimethoxy-3,4-dihydro-1H-isochromen-1-one (7) (1.5g,

6.3mmol) was dissolved in dry THF (20mL) and treated with magnesium (0.2g, 7.58

mmol) and iodine 1.0g, 8.38 mmol) in dry benzene (30mL). The resulting mixture was

refluxed for 30min. and then poured into water and organic layer was separated and

washed with water. Evaporation of the solvent under reduced pressure afforded 8-

hydroxy-7-(hydroxymethyl)-6-methoxy-3,4-dihydro-1H-isochromen-1-one (8), (Yield

111

47%); Rf: 0.5 (petroleum ether and ethyl acetate 4:1); m. p. 127-128 °C; IR (KBr): 3559

(O-H), 3001 (C-H), 2920 (Ar-C-H), 1710 (C=O), 1593 (C=C) cm-1

; 1H NMR (CDCl3, δ

ppm ): 10.90 (1H, s, 8-OH), 7.06 (1H, s, H-5), 4.54 (2H, s, CH2-OH), 4.12 (2H, t, J=3.7,

H-3), 3.90 (3H, s, 6-OCH3), 2.86 (2H, t, J=3.7, H-4), 2.29 (1H, s, CH2O-H); 13

C NMR

(CDCl3, δ ppm): 166.5 (C1), 158.7 (C8), 151.4 (C6), 143.1 (C4a), 116.9 (C7), 105.2

(C8a), 102.6 (C5), 63.9 (C3), 56.9 (6-OCH3), 47.1 (CH2-OH), 28.4 (C4); MS (70eV): m/z

(%); 224 [M]+.

(59), 222 (21), 207 (42), 194 (100), 30 (19); Anal. Calcd. for C11H10O5 C,

59.45 H, 4.50 Found C, 59.37 H, 4.39 O.

5.2 Results and Discussion

Vilsmeier Haack formylation of the acetate (1) with phosphorous oxychloride in

N, N-dimethylformamide (DMF) afforded the methyl (2-formyl-3, 5-dimethoxy-4-methyl

phenyl) acetate (2). The IR showed very strong new carbonyl absorption at 1690 cm-1

for

aldehydic carbonyl in addition to the 1722 cm-1

peak for ketonic carbonyl already

present. The 1H NMR showed the singlet for aldehydic proton at δ 9.75 ppm and the

characteristic changes in the chemical shifts of the benzylic protons. 13

C NMR showed a

peak at δ 179.3 ppm for aldehydic carbon and a peak at 162.4 for ester carbon already

present. The molecular ion peak appeared at m/z 252 with 25% bundance and the base

peak at m/z 165.

The aldehyde (2) was oxidized to 2,4-Dimethoxy-6-(2-methoxy-2-oxoethyl)-3-

methylbenzoic acid (3) by using sulfamic acid and sodium chlorite at 0°C in 79% yield.

The carbonyl absorption in IR shifted from 1690 cm-1

to 1715 cm-1

due to oxidation of

aldehydic function into carboxyl one. The absorption at 3265 cm-1

for (O-H) is also

present in IR spectrum. In 1H NMR a singlet at δ 8.19 ppm appeared for carboxylic

proton and downfield shift from δ 179.3 to δ 197.7 ppm for carboxylic carbon was also

appeared in 13

C NMR.

The ester acid (3) was then reduced to 2,4-dimethoxy-6-(2-hydroxyethyl)-3-

methylbenzoic acid (4) using sodium borohydride-methanol system refluxed in freshly

distilled THF. The IR showed a broad band at 3481 cm-1

for the hydroxyl group and also

the disappearance of ester carbonyl absorption was noticed. In 1H NMR a singlet for

hydroxyl proton appeared at δ 2.03 ppm and two methylenes showed two triplet at δ 4.23

ppm, J=3.8Hz and δ 2.65 ppm, J=3.8Hz. The reduction of esters and similar functional

112

groups using sodium borohydride is relatively difficult to obtain and it has not been

widely used. However, the reactivity of the sodium borohydride can be enhanced by

carrying out the reaction in the presence of NaBH4-CH3OH. This methodology is simple,

safe, inexpensive, and general and the reduction of methyl esters was completed after

refluxing in THF.

Then cyclization of the alcohol acid (4) was carried out in the presence of acetic

anhydride refluxed in dry toluene to afford 6,8-dimethoxy-7-methyl-3,4-dihydro-1H-

isochromen-1-one (5). In IR spectrum the absorption band for hydroxyl disappeared and

the lactonic carbonyl showed absorption at 1702 cm-1

. Similarly in 1H NMR the singlet

for hydroxyl and carboxyl protons were also disappeared. The lactonic carbonyl carbon

showed peak at δ 163.9 ppm in 13

C NMR spectrum.

OCH3

H3CO

CH3

O

OH

CH3OH

Conc.H2SO

4 OCH3

H3CO

CH3

O

OCH3

OCH3

H3CO

CH3 COOH

OHNaBH

4/ THF

CH3OH

(1)

(4)

O

O

CH3

OCH 3

H3CO

Acetone/H2O Mg/I2

THF/Benzene

O

O

CH2

OCH3

H3CO

Br

(5) (6)

O

O

CH2

OCH3

H3CO

OH

(7)

O

O

CH2

OH

H3CO

OH

(8)

(CH3CO)

2O

1h reflux

CH3

OCH3

H3CO O

OCH3O

H

(2)

CH3

OCH3

H3CO O

OCH3O

OH

(3)

POCl3 / Freshly distilled DMF

CH3COO- Na

+

NH2SO3H / NaClO2 / 0 °C

H2O:THF:DMSO

Benzoylperoxide /CCl4

N-bromosuccinimide

Scheme 5.1 Synthesis of Stellatin

113

The benzylic bromination of the 6,8-dimethoxy-7-methyl-3,4-dihydro-1H-

isochromen-1-one (5) in the presence of N-bromosuccinimide and catalytic amount of

benzoyl peroxide refluxed in dry CCl4 afforded 6,8-dimethoxy-7-(bromomethyl)-3,4-

dihydro-1H-isochromen-1-one (6). In 1H NMR, the singlet for 7-methyl hydrogens

shifted downfield after bromination from δ 2.66 ppm to δ 4.85 ppm. Then 6,8-dimethoxy-

7-(bromomethyl)-3,4-dihydro-1H-isochromen-1-one (6) was converted into 7-

(hydroxymethyl)-6,8-dimethoxy-3,4-dihydro-1H-isochromen-1-one (7) by nucleophilic

substitution using acetone and H2O. IR showed a broad band at 3467 cm-1

for hydroxyl

group. In 1H NMR, a singlet for hydroxyl proton appeared at δ 2.36 ppm.

The selective demethylation of the 8-hydroxyl group in 7-(hydroxymethyl)-6,8-

dimethoxy-3,4-dihydro-1H-isochromen-1-one (7) using Mg/I2 refluxed in THF/benzene

afforded 8-hydroxy-7-(hydroxymethyl)-6-methoxy-3,4-dihydro-1H-isochromen-1-one

(Stellatin) (8). In 1H NMR, a singlet at δ 10.90 ppm appeared for 8-hydroxy and 7-

hydroxy proton appeared as a singlet at δ 2.29 ppm. The physical constants and FTIR

spectral data of the compounds (1-8) are presented in Table 5.1.

Table 5.1 Physical and FTIR spectral data of the compounds (1-8)

Compds M. P.

(°C)

Rf Yield

(%) Ar-C-H Sp

3 C-H C=O C=C O-H

1 38-40 0.7 87 3023 2925 1734 1583 -

2 51-53 0.55 85 3015 2917 1722,

1690 1595 -

3 164-166 0.4 79 3011 2928 1734,

1715 1582 3265

4 72-174 0.3 83 3009 2924 1710 1574 3481,

3034

5 145-147 0.65 78 3010 2935 1702 1591 -

6 91-93 0.6 81 3013 2926 1718 1582 -

7 136-138 0.4 77 3010 2919 1714 1587 3467

8 127-128 0.5 45 3001 2920 1710 1593 3559,

3178

Pet. Ether: Ethyl Acetate (4:1)

114

Table 5.2 1H and

13C NMR data of Stellatin (8)

Carbons δ (ppm) and multiplicity

1H NMR

13C NMR

C-1 ----- 166.5

C-3 4.12, t, J=3.7Hz 63.9

C-4 2.86, t, J=3.7Hz 28.7

C-4a ----- 143.1

C-5 7.06, s 102.6

C-6 ----- 15.4

C-7 ----- 116.9

C-8 ----- 158.7

C-8a ----- 105.2

6-OCH3 3.90, s 56.9

7-CH2 4.56, s 47.1

8-OH 10.90, s -----

CH2-OH 2.29, s -----

The formation of the 8-hydroxy-7-(hydroxymethyl)-6-methoxy-3,4-dihydro-1H-

isochromen-1-one (8) was confirmed by the presence of two triplets at δ 4.12 ppm with

coupling constant 3.7Hz for H-4 and 2.86 ppm with coupling constant 3.7Hz for H-3

hydrogens in 1H NMR spectrum. A singlet at δ 3.90 ppm appeared for 6-methoxy

hydrogens. The 8-hydroxy appeared as a singlet at δ 10.90 ppm in 1H NMR spectrum. It

was also supported in 13

C NMR spectrum by the presence of a peak at δ 166.5 ppm for

lactonic carbon C-1. The detailed 1H and

13C NMR data of the compound (8) are

presented in Table 5.2.

115

The structure of the Stellatin (8) was further confirmed by mass spectrometry.

The molecular ion peak appeared at m/z 224 with 59% abundance which confirmed the

formation of isocoumarin (8). By the removal of hydroxyl radical from molecular ion, a

peak at m/z 207 with 42% abundance appeared. A peak at m/z 194 is the base peak

formed by the elimination of formaldehyde fragment from molecular ion. The

fragmentation pattern of the Stellatin (8) is shown in Fig. 5.1.

m/z = 224 [M]+.

59%m/z = 207, 42%m/z = 30, 19%

m/z = 179, 51%

CO_

_

CH2

O

H3CO

OH

OH

m/z = 194, 100%

m/z = 196, 47%

+.

+

+

+

H3CO

OH

OOH

O

O

H3CO

OHOH

-OH.

+

O

O

H3CO

OH

+

O

H3CO

OH

O

OH

H3CO

OH

CO_

.

CH2O

.

CH2

O

_.

m/z = 206, 63%

+.

O

O

H3CO

O

-H2O

-H2

m/z = 222, 21%

+.

O

O

H3CO

OO

Fig. 5.1 Mass fragmentation pattern of the Stellatin (8)

116

5.3 Antibacterial Activity

Bacterial infections constitute one the most serious situations in infectious

disease. The detection and identification of these bacteria is one of the most important

functions of clinical microbiology. Isolation of an infectious agent from the patient with

disease is often not sufficient for determining proper therapy. Since the susceptibility of

many bacteria to antimicrobial agents cannot be predicted testing individual pathogens,

against appropriate agent (with the most activity against the pathogen, the least toxicity to

the most, the least important on normal flora, appropriate pharmacologic characteristics

and most economical) can then be chosen allowing a more certain therapeutic outcome.

Antibacterial activity of the compounds (5-8) was determined against various

gram positive and gram negative bacterial strains by using agar well diffusion. The

purified samples were dissolved in DMSO 5mg/ml. DMSO is the negative control and

antibiotic chloramphenicol is the positive control in this invitro antibacterial study.

Ten bacterial strains Escherichia coli (E. c.), Klebsiella pneumonae (K. p.),

Lactobacillus bulgaricus (L. b.), Micrococcus luteus (M. l.), Pasteurella multocida (P.

m.), Proteus vulgaris (P. v.), Pseudomonas aeruginosa (P. a.), Salmonella typhi (S. t.),

Staphylococcus aureus (S. a.) and Staphylococcus epidermidis (S. e.) were selected in

this antibacterial assay. Micrococcus luteus, Staphylococcus aureus and Staphylococcus

epidermidis are the example of Gram positive and the remaining seven are gram negative

bacteria. All of the tested microorganisms were maintained on nutrient agar at 4°C and

sub-cultured before use. The bacteria studied are clinically important ones causing

several infections and it is essential to overcome them through some active therapeutic

agents.

The antibacterial assay was performed by agar well diffusion method against

different bacterial strains. Each tested bacterium was sub-cultured in nutrient broth at

37°C for 24h. One hundred micro liters of each bacterium was spread with the help of

sterile spreader on to a sterile Muller-Hinton agar plate so as to achieve a confluent

growth. The plates were allowed to dry and wells (6mm diameter) were punched in the

agar with the help of cork borer. 0.1mL of the each compound solution (5mg/mL) in

DMSO was introduced in to the well and the plates were incubated overnight at 37°C.

117

The antimicrobial spectrum of the compounds was determined for the bacterial

species in terms of size of the zones around each well. The diameters of the zone of

inhibition produced by the compounds were compared with those produced by the

commercial antibiotic chloramphenicol (5mg/mL). This is the common antibiotic used

for the treatment of infections caused by gram positive and gram negative bacteria. The

control activity was deducted from the test and the results obtained were plotted. The

experiment was performed three times to minimize the error and the mean values are

presented.

Anti bacterial activity results of the compounds (5-8) is shown in table 5.3

respectively.

Table 5.3 In vitro Antibcterial activity of compounds (5-8)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

5 3 1 0 11 0 0.5 1 3 10 11

6 1 7 2 9 10 0 1.5 2 6 7

7 11 7 5 0.5 8 1 3 0 0.5 1.5

8 14 8 9 1 11 6 2 12 1 3

Standard 18 10 13 13 12 13 13 14 13 13

*Activity of each sample is measured by subtracting the activity of DMSO.

Escherichia coli (E. c.), Klebsiella pneumonae (K. p.), Lactobacillus bulgaricus (L. b.),

Micrococcus luteus (M. l.), Pasteurella multocida (P. m.), Proteus vulgaris (P. v.),

Pseudomonas aeruginosa (P. a.), Salmonella typhi (S. t.), Staphylococcus aureus (S. a.)

and Staphylococcus epidermidis (S. e.)

118

5.4 Cytotoxic Activity

The Neutralrottest was carried out according to the protocol of the National

Institutes of Health (NIH). In the vital dye neutral red is taken up by living cells and then

protonated. The cell acts as an ion trap, allowing the dye can not diffuse out of it. By

destroying the cells, the neutral is released again and can be determined photometrically.

The absorption is a measure of cell viability, the lower the absorption were the less living

cells. For the tests, the immortalized human Keratinocyte cell line (HaCaT) was used.

The incubation with the test substances was carried out over three days and the

experiments were conducted in two independent experiments with a number of parallels.

The stock solutions of substances were prepared in DMSO. The concentration of the

solvent DMSO was all tested concentrations of 0.1%. Etoposide as positive control was

carried in a concentration of 10μM (IC50). Concentration range of the tested samples is

1.56-100 (µM).

Fig. 5.2 Cytotoxic activity of the samples (5-8)

The results showed that cytotoxic activity increases as the concentration of the

samples increases. The cytotoxic activity of the final stellatin is higher as compared to the

precursors. Compounds 5 and 6 showed moderate to low cytotoxic activity. When 7-

methyl group in compound 5 is converted into 7-hydroxymethyl group in compound 7,

cytotoxic activity is increased. After selective demethylation of the 8-methoxy group in

compound 8 results in increase of cytotoxic activity of the final compound stellatin.

Presence of the 7-hydroxymethyl and 8-hydroxy functional groups play vital role in

cytotoxic activity of the final compound.

0

20

40

60

80

100

120

10 25 50 100 1000

Via

bilit

y [

%]

Concentration [uM]

5

6

7

8

119

6. REFERENCES PART I

1. Bu’Lock, J. D. The Biosynthesis of Natural Product, an introduction to secondary

metabolite. Mcgraw-Hill, New York, London, 1965.

2. Barry, R. D. Chem. Rev.1964, 64, 229.

3. Turner, W. B.; Aldridge, D. C. Fungal metabolites II; 1983, Academic Press,

London.

4. Yamato, V.; Yuki Gosei kagaku kyokaishi., 1983, 41, 958.

5. Hill, R. A. Progress in the Chemistry of Organic Natural Products. 1986, 49, 1-

78.

6. Napolitano, E. Organic Preparations and Procedures Int., 1997, 26, 631.

7. Bin, Y.; Song, L.; Xiaohui, G. Tianran Chanwu Yanjiu Yu Kaifa., 2000, 12, 95.

8. Filho, R. B.; De Moraes, M. P. L.; Gottieb, O. R. Phytochem. 1980, 19, 2003.

9. Vogel, A. Gilbert’s Ann. Phy., 1820, 64,161.

10. Henderson, G. B.; Hill, R. A. J. Chem. Soc. Perkin Trans. I., 1982, 1111.

11. Cantello, B. C. C.; Buckle, D. R.; Smith, H. UK patent, GB1480737, 1977.

12. Bailey, D. M.; DE Grazia, C. G. J. Org. Chem., 1970, 35, 4088.

13. Turner, W. B. Fungal metabolites, 1971, Academic Press London.

14. Suzuki, Y. Agric. Biol. Chem; (Japan), 1970, 34, 760.

15. Mcinerney, B. V.; Taylor, W. C.; Lacey, R. J.; Akhurst, R. J.; Gregson, R. P. J.

Nat. Prod., 1991, 785-795.

16. a) Curtis, R. F.; Hassall, C. H.; Nazer, M. J. Chem. Soc. (C), 1968, 85. b) Barber,

J.; Garson, J. Stanton, J. J. chem. Soc., Perkin Trans. 1, 1981, 2584. c) Fujii, I.;

Watanebe, A.; Sankawa, U.; Ebiauka, Y. Chem. Biol., 2001, 8, 189.

17. Maneekarn, C.; Prasat, K.; masahiko, I.; Ratchanda, C.; Morako, T.; Yodhathai,

T. J. chem. Soc., Perkin Trans. 1, 2002, 2473-2476.

18. Hiroyuki, K.; Masahide, A.; Hiroshi, N,; Tsutomu, S.; masaaki, I.; Tomio, T.

J.Antibiotics, 1994, 47, 4, 440-446.

19. Furutani, Y.; Tsuchiya, I.; Naganawa, H.; Takeuchi, T.; Umezawa, H. Agric. Biol.

Chem. 1977, 41, 1581-1585.

20. Christopher, N. L.; James, S.; David, C. S. J. chem. Soc., Perkin Trans. I, 1988,

747-754.

120

21. De Jesus, A. E.; Steyn, P. S.; Vieggaar, R.; Wessels, P. L. J. chem. Soc., Perkin

Trans. 1, 1980, 52.

22. Wiesleder, D.; Lillehoj, E. Tetrahedron Lett., 1980, 21, 993.

23. Jonathan, P. H.; Peter, G. M. Phytochemistry, 2001, 58, 709-716.

24. Matsuda, H.; Shimoda, H.; Yoshikawa, M. Bioorg. Med. Chem., 1999, 7, 1445-

1450.

25. Yoshikawa, M.; Harada, E.; Naitoh, Y.; Inoue, K.; Matsuda, H.; Shimoda, H.;

Yamahara, J.; Murakami, N. Chem. Pharm. Bull., 1994, 42, 2225-2230.

26. a) Matsuda, H.; Shimoda, H.; Yamahara, J.; Yoshikawa, M.; Bioorg. Med. Chem.

Lett., 1998, 8, 215-220. b) Shimoda, H.; Matsuda, H.; yamahara, J.; Yoshikawa,

M.; Biol. Pharm. Bull., 1998, 21, 809-813.

27. Nozawa, K.; Yamada, M.; Tsuda, Y.; kawai, K.; Nakajima, S.; Chem. Pharm.

Bull., 1981, 29, 2689-2691.

28. Furuta, T.; Fukuyama, Y.; Asakawa, Y. Phytochemistry, 1986, 25, 517-520.

29. Whyte, A. C.; Gloer, J. B.; Scott, J. A.; Mallock, D. J. Nat. Prod., 1996, 59, 765-

769.

30. Lee, J. H.; Park, Y. J.; Kim, H. S.; Hong, Y. S.; kim, K. W.; Lee, J. J.; J. Antibiot.,

2001, 54, 463-466.

31. Bloomquist, J. R. Arch. Insect Biochem. Physiol., 2003, 54, 145-156.

32. Yoshihisa, O.; Tadahiko, K.; Yuji, T.; Kenzo, H.; Nobutoshi, T.; Takashi, Y.;

Emiko, S.; Kei-ichi, I.; Kazuhiko, O. J. Pestic. Sci., 2004, 29, 4, 328-331.

33. Zhi-Hong, X.; Li-Tian,; Tian-jiao, Z.; Wen-Liang, W.; Lin, D.; Yu-chun, F.;

Qian-Qun, G, Wei-Ming, Z. Arch. Pharm. Res. 2007, 30,7, 816-819.

34. Yong-Fu, H.; Lin-Hao, L.; Li, T.; Li, Q.; Hui-Ming, H.; Yue-Hu, P. J. Antibiot.

2006, 59, 6, 355-357.

35. Shimojima, Y.; Hayashi, H.; Ooka, T.; Shibukawa, M. Agric. Biol. Chem., 1982,

46, 1823-1829.

36. Itoh, J.; Omoto, S.; Shomura, T.; Nishizawa, N.; Miyado, S.; Yuda, Y.; Shibata,

U.; Inouye, S. Agric. Biol. Chem., 1982, 46, 1255-1259.

37. Mcinerney, B. V.; Taylor, W. C.; Lacey, M. J.; Akhurst, R. J.; Gergson, R. P. J.

Nat. Prod., 1991, 54, 785-795.

121

38. Tutomu, S.; koji, N.; Kenichi, S.; Moto, M.; Takeshi, S. The Journal of

Antibiotics, 1992, 45, 12, 1949-1952.

39. Rodrigues, K. F.; Petrini, O. Biodiversity of Tropical Microfungi (ed. K. D. Hyde),

Hong Kong, 1997, 57-69.

40. Lin, Y.; wan, J.; Zhou, S.; Gareth, J. CJI, 2001, 3, 7, 30.

41. Razieh, Y.; Hamid, R. A.; Ataback, B. DARU, 2000, 8, 1 & 2, 42-44.

42. Danise, C. E.; Keller, G. G.; Tamara, P. K.; john, M. P.; Fernao, C. B. J. Nat.

Prod., 2008, 71, 6, 1082-1084.

43. Karina, F. D.; Maria, S. G. R.; Eliana, A. V.; wagner, V. Z. Naturforsch., 2002,

57c, 85-88.

44. Kithsiri, E. M. W.; Priyani, A. P.; Leslie, A. A. G. Tetrahedron, 2006, 62, 34,

8439-8446.

45. Perreault, S. D.; Zirkin, B. R. J. Exp. Zool., 1982, 224, 253-257.

46. Tummon, I. S.; Yuzpe, A. A.; Daniel, S. A.; Duetsch, A. Fertil. Steril., 1991, 56,

933-938.

47. Francavilla, S.; Palermo, G.; Gabriele, A.; Cordeschi, G.; Poccia, G. Fertil. Steril.,

1992, 57, 1311-1316.

48. De Jonge, C. J.; Tarchala, S. M.; Rawlins, R. G.; Binor, Z.; Radwariska, E. Hum.

Reprod., 1993, 8, 253-257.

49. Palencia, D. D.; Garner, D. L.; Hudig, D.; Holcombe, D. W.; Burner, C. A.;

Redelman, D.; Fernandez, G. C. J.; Abuelyaman, A. S.; Kam, C. M.; Powers, J. C.

Biology of Reproduction, 1996, 55, 536-542.

50. Kumagai, H. et al., Journal of Antibiotics, 1990, 72, 1505.

51. Shin-ichi, H.; Toshiyuki, M.; Naoki, A.; Hiroshi, I.; Naoki, M.; Takeo, Y.;

Hiroshi, T.; Hiroyuki, K.; Massaki, I.; Tomio, T. US Pat. Appl., 2000, US

6020363.

52. Neidleman, S. L.; Geigert, J. Biohalogenation, 1986, 13-15.

53. Marc, S.; Heidrun, A. The Journal of Antibiotics, 1995, 48, 3, 261-266.

54. Kongsaeree, P.; Prabpai, S.; Sriubolmas, N.; Vongvein, C.; Wiyakrutta, S. J. Nat.

Prod., 2003, 66, 5, 709-711.

122

55. Oikawa, T.; Sasaki, M.; Inose, M.; Shimamura, M.; Kobuki, H.; Hirano, S.;

Kumagai,H.; Ishizuka, M.; Takeuchi, T. Anticancer Res., 1997, 17, 1881-1886.

56. Nakashima, T.; Hirano, S.; Agata, N.; Kumagai, H.; Isshiki, K.; Yoshioka, T.;

Ishizuka, M.; Maeda, K.; Takeuchi, T. J. Antibiot., 1999, 52, 426-428.

57. Reimer, C. L.; Agata, N.; Tammam, J. G.; Bamberg, M.; Dickerson, W. M.;

Kamphaus, G. D.; Rook, S. L.; Milhollen, M.; Fram, R.; Kalluri, R.; Kufe, D.;

Kharbanda, S. Cancer Res. 2002, 62, 789-795.

58. Agata, N.; Nogi, H.; Bamberg, M.; Milhollen, M.; Pu, M.; Weitman, S.;

Kharbanda, S.; Kufe, D. Cancer Chemother. Pharmacol. 2005, 56, 610-614.

59. Kunihiro, I.; Yohei, M.; Yoshihiko, Y.; Masaru, K.; Kumiko, H.; Hiroyuki, K.;

Yuki, T.; Hitoshi, S.; Yasushi, Y.; Naoki, A.; Hirofumi, M. Diabetes, 2006, 55,

1232-1242.

60. Agata, N.; Nogi, H.; Milhollen, M.; Kharbanda, S.; Kufe, D. Cancer Res. 2004,

64, 8512-8516.

61. Wang, Y. Med. Res. Rev., 2001, 21, 146-170.

62. Romer, J.; Neilsen, B.; Ploug, M. Curr. Pharm. Des., 2004, 10, 2359-2376.

63. Jo, M.; Thomas, K.; Marozkina, N.; Amin, T.; Silva, C.; Parsons, S.; Gonias, S. J.

Biol. Chem., 2005, 280, 17449-17457.

64. Justin, J. H.; Lucy, A. H.; Thomas, A. V. J.; Lorraine, M. D.; David, L. V. J. BMC

Chemical Bilogy, 2006, 6, 1472.

65. Narasimhan, N. S.; Mali, R. S. Topics in current chemistry, 1987, 138, 63.

66. Woon, E. C. Y.; Dhami, A.; Mahon, M. F.; Threadgill, M. D. Tetrahedron.

2006, 62, 4829.

67. Subramanian, V.; Batchu, V. R.; Barange, D.; Pal, M. J. Org. Chem. 2005,

70, 4778.

68. Roy, H.; Sarkar, M. Synth. Commun. 2005, 35, 2177.

69. Cherry, K.; Parrain, J. L.; Thibonnet, J.; Duchene, A.; Abarbri, M. J. Org. Chem.

2005, 70, 6669.

70. Suzuki, T.; Yamada, T.; Watanabe, K.; Katoh, T. Bioorg. Med. Chem. Lett.

2005, 15, 2583.

71. Opatz, T.; Ferenc, D. Eur. J. Org. Chem. 2005, 817.

123

72. Martinez, A.; Fernandez, M.; Estevez, J. C.; Estevez, R. J.; Castedo, L.

Tetrahedron 2005, 61, 485.

73. Yao, T.; Larock, R. C. J. Org. Chem., 2003, 68, 5936.

74. Liao, H.-Y.; Cheng, C. H. J. Org. Chem., 1995, 60, 3711.

75. Bonadies, F.; DiFabio, R. J. Org. Chem., 1984, 49, 1647.

76. Kinder, M. A.; Kopf, J.; Margaretha, P. Tetrahedron, 2000, 56, 6763.

77. Colonge, J.; Boisde, P. Bull. Soc. Chem. France, 1956, 1337.

78. Carter, R. H.; Colyer, R. M.; Hill, R. A.; Staunton, J. J. Chem. Soc. Perkin. Trans.

I., 1982, 1438.

79. Kendall, J. K.; Fisher, T. H. J. Org. Chem., 1989, 54, 4218.

80. Mal, D.; Bandyopadhyay, M.; Datta, K.; Murty, K. V. S. N. Tetrahedron, 1998,

54, 7525.

81. Mal, D.; Bandyopadhyay, M.; Sujit, K.; Datta, K. Tetrahedron Lett., 2000, 41, 1.

82. Kinder, M. A.; Kopf, J.; Margaretha, P. Tetrahedron, 2000, 56, 6763.

83. a) Vaulx, R. L.; Puterbauth, W. H.; Hauser, C. R. J. Org. Chem., 1964, 29, 3514.

b) Mao, C. L.; Barnish, I. T.; Hauser, C. R. J. Heterocyle. Chem., 1969, 6, 83.

84. Narasimhan, N. S.; Bhide, B. H. Tetrahederon, 1971, 27, 6171.

85. Narasimhan, N.S.; Mali, R. S. Synthesis, 1983, 63.

86. Snieckus, V. Chem. Rev., 1990, 90, 879.

87. Lee, D.; Still, W.C. J. Org. Chem., 1989, 54, 4715.

88. Reitz, D. B.; Massey, S. M. J. Org. Chem., 1990, 55, 1375.

89. Superchi, S.; Minutolo, F.; Pini, D.; Salvadori, P. J. Org. Chem., 1996, 61, 3183.

90. Mroady, S. M.; Rexhausen, J. E.; Thomas, E. J. J. Chem. Soc. Perkin Trans. 1.,

1999, 1083.

91. Pini, D.; Superchi, S.; Salvadori, P. J. Organometallic. Chem., 1993, 452.

92. Gruniwald, G. L.; Dahanukar, V. H. J. Heterocyclic Chem., 1994, 31, 1609.

93. Bestmann, H. J.; Kern, F.; Schafer, D.; Witschel, M. C. Angew. Chem., Int. Ed.

Engl., 1992, 31, 795.

94. Choukchou-Braham, N.; Asakawa, Y.; Lepoittevin, J. P. Tetrahedron Lett., 1994,

35, 3949.

95. Bhide, B. H.; Akolkar, V. D.; Brahmbhat, D. I. Ind. J. Chem., 1992, 31(B), 116.

124

96 Kurosaki, Y.; Fukuda, T.; Iwao, M. Tetrahedron, 2005, 61, 3289–3303.

97. Zenner, J. M.; Larock, R.C. J. Org. Chem., 1999, 64, 7312.

98. Kessar, S. V.; Singh, P.; Vohra, R.; Kaur, N. P.; Venugopra, D. J. Org. Chem.,

1992, 57, 6716.

99. Conners, R.; Tran, E.; Durst, T. Can. J. Chem., 1996, 74, 221.

100. Larock, C. L.; Varaprath, S. US Pat. Appl., 1987, US 4650881.

101. Nagarjaan, A.; Balasubramanian, R.T. Indian J. Chem., Sect.B, 1987, 26B, 917.

102. Roy, H. N.; Sarkar, M. S. Synthetic Communications, 2005, 35, 2177.

103. Stephen, P. W.; Marisa, C. K. Tetrahedron Letters., 2001, 42, 3567-3570.

104. Suzuki,T.; Yamada, T.; Watanabe, K.; Katoh, T. Biorg. Med.Chem. Lett. 2005,

15, 2583-2585.

105. Ueura, K., Satoh, T., Miura, M., J. Org. Chem. 2007, 72, 5362.

106. Loewenthal, H. J. E.; Pappo, R. J. Chem. Soc., 1992, 4799.

107. Chatterjea, J. N.; Mukherjea, H. Experietia, 1960, 16, 439.

108. Chatterjea, J. N.; Mukherjee, H. J. Indian chem. Soc., 1960, 37, 379.

109. Chatterjea, J. N.; Mukherjee, H. J. Indian chem. Soc., 1960, 37, 443.

110. Yamato, M.; Hashigaki, K. Chem. Pharm. Bull., 1976, 24, 200.

111. Kabayashi, T. Sci. Rept. Tohoku Univ., First Ser., 1942, 31, 73; C. A., 1950, 44,

4013.

112. Haworth, R. D.; Pindred, H. K.; Jafferies, P. R. J. Chem. Soc., 1954, 3617.

113. Chatterjea, J. N. J. Indian chem. Soc., 1953, 30, 103.

114. Vorozhtsov, N. N.; Petushova, A. T. J. Gen. Chem. USSR, 1957, 27, 2282.

115. Tirdkar, R. B.; Usgoankar, R. N. Indian J. Chem., 1970, 8, 123.

116. Modi, R.; Usgoankar, R. N. Indian J. Chem., 1979, 17B, 360.

117. Rose, A.; Buu-Hoi, N. P.; Jacquinon, P. J. Chem. Soc., 1965, 6100.

118. Yoshikawa, H.; Taniguchi, E.; Maekawa, K. J. Pesticide Sci., 1980, 5, 1.

119. Sarkhel, B. K.; Srivasta, J. N. J. Indian chem. Soc., 1976, 53, 915; Ibid., 1977, 54,

925.

120. Tuanli Yao and Richard C. Larock, Tetrahedron Lett., 2002, 43, 7401.

121. Berti, G. Tetahedron, 1958, 4, 393.

122. Muller, E. Chem. Ber., 1909, 42, 423.

125

123. Ribbens, C.; Koninkl, N. V.; Fabrieken, P. v/h brocades-Stheeman Pharmacia,

1960-61, 10, 9. C. A. 1962, 56, 7378.

124. Stadlbauer, W.; Ghobrial, N.; Kappe T. Z. Naturforsch, 1980, 35b, 892.

125. Prey, V.; Kerres, B.; Berbalk, H. Monatsh. Chem., 1960, 91, 774.

126. Berti, G.; Marsili, A.; Mini, V. Ann. Chim.(Rome), 1960, 50, 669.

127. Birk, A. J.; Donovan, F. W. Australian J. Chem., 1953, 6, 360.

128. Colonge, J.; Boisde, P. Bull. Soc. Chim. France, 1956, 1337.

129. Maitte, P. Compt. Rend., 1954, 239, 1508.

130. Siegel, S.; Colburn, S. K.; Levering, D. R. J. Am. Chem. Soc., 1951, 73, 3163.

131. Shriner, R. L.; Knox, W. R. J. Org. Chem., 1951, 16, 1064.

132. Alder, E.; Magnusson, R.; Berggren, B. Acta Chem, Scand., 1960, 14, 539.

133. Grimshaw, J.; Haworth, R. D.; Pindred, H. K. J. Chem. Soc., 1955, 833.

134. Yamamoto, I. Agri. Biol. Chem. (Tokyo), 1961, 25, 400; C. A., 1961, 55, 670.

135. Thomas, O.L.; Jens, B. J. Nat. Prod., 1999, 62, 1182.

136. Stadler, M.; Anke, H.; Sterner, O. J. Antibiot., 1995, 48, 261.

137. Yamato, M.; Hashigaki, K.; Honda, E.; Sato, K.; Koyama, T. Chem. Pharm. Bull.,

1977, 25, 695.

138. Liu, D. Zhonghua Yixue Zashi, 1982, 62, 336.

139. Haworth, R. D.; Pindred, H. K.; Jafferies, P. R. J. Chem. Soc., 1954, 3617.

140. Nozawa, K.; Yamada, M.; Tsuda, Y.; Kawai, K. I.; Nakajima, S. Chem. Pharm.

Bull., 1981, 29, 2491.

141. Berti, G. J. Org. Chem., 1959, 24, 934.

142. Okeke, M. I.; Iroegbu, C. U.; Eze, E. N.; Okoli, A. S.; Esimone, C. O. Journal of

Ethnopharmacology, 2001, 78, 119-127.

143. Plowe, C. V.; Djimde, A.; Bouare, M.; Doumbo, O.; Wellens, T. E. Am. J. Trop.

Med. Hyg. 1995, 52, 6, 565-568.

144. Omulokoli, E.; Khan, B.; Chhabra, S. C. J. Ethnopharmacol. 1997, 56, 133-137.

126

7.1 INTRODUCTION

Isocoumarins (1H-2-benzopyran-1-ones) are the secondary metabolites of an

extensive variety of fungi, bacteria, higher plants, marine organisms and are also

among insect venoms and pheromones; exhibiting a wide range of structural diversity and

biological activities1-3

. Important examples of natural bioactive isocoumarins include

the furoisocoumarin coriandrin phototoxic to RNA-virus Sindbis virus, DNA-virus

murine cytomegalo-virus and HIV4, thunberginol, phyllodulcin and hydrangenol having

differentiation inducing, antiallergic, and immunomodulatory effects5, ochratoxins A &

B, nephratoxic, hepatotoxic6, hiburipyranone, cytotoxic

7, duclauxin, antitumor

8,

cytogenin and its synthetic analogues antitumor, antidiabetic anticancer9 and Sg17-1-4,

possessing potent cytotoxic activities10

.

Majority of the natural isocoumarins being of polyketide origin are derived

biogenetically from acetate-polymalonate pathway, hence most of them possess a C-3

alkyl / aryl substituent. Although, more than two hundred isocoumarins and

dihydroisocoumarins have been isolated and the number is still increasing dramatically,

1-thioisocoumarins are thus far unknown in nature. A review of literature reveals that

while, the thio analogues of a number of associated natural products viz., chromones,11

flavones,12

isoflavones13

and coumarins14

have been prepared, the reports of synthetic 1-

thioisocoumarins (1H-isochromene-1-thiones) are exceptional15

.

1(2H)-isoquinolones (isocarbostyrils) are the nitrogen analogues of isocoumarins

(1H-2-benzopyran-1-ones). Various 1(2H)-isoquinolone derivatives are found in several

bioactive natural products such as thalifoline, doryphorine16

, uprechstyril

17, narciclasine

18,

pancratistatin, lycoricidine19

, the alkaloids coryaldine20

, dorianine21

hydroxyhydrastinine

and thalflavine21

. Isoquinolone nucleus is also an integral part of complex isoquinoline

alkaloids and is a useful building block in organic synthesis.

The isoquinolone skeleton biogenetically derived from amino acid phenylalanine,

exhibits biomimetic characteristics22

. Substituted isoquinolones are orally effective

antagonists of receptors 5-HT3, which have shown higher efficacy in the control of cancer

models23

, thymidylate synthase (TS) inhibitors24

, human Tumor Necrosis Factor (TNF)

inhibitors, and tachykinin receptors25

. Substituted isocarbostyrils showing

127

antidepressant26

, anti-inflammatory27

, analgesic28

and hypolipidemic29

activities have also

been reported.

7.2 EXPERIMENTAL

Melting points were recorded using a digital Gallenkamp (SANYO) model MPD

BM 3.5 apparatus and are uncorrected. 1H NMR spectra were determined as CDCl3

solutions at 300 MHz on a Bruker AM-300 spectrophotometer. FT IR spectra were

recorded using an FTS 3000 MX spectrophotometer; Mass Spectra (EI, 70eV) on a GC-

MS instrument and elemental analyses with a LECO-183 CHNS analyzer. All

compounds were purified by thick layer chromatography using silica gel from Merck.

Synthesis of homophthalic anhydride (1)

A solution of homophthalic acid (2.0g, 12.34 mmol) in dry toluene (35 mL) was

treated with acetic anhydride (1.1g, 10mL, 10.8 mmol). The reaction mixture was

refluxed for 1 hr and then poured into ice cold water. The organic layer was separated,

dried over anhydrous sodium sulfate and toluene was rotary evaporated to get

homophthalic acid anhydride (1). Yield 82%; Rf: 0.7 (petroleum ether and ethyl acetate,

4:1); m. p. 140-142°C; IR (KBr): 3011 (C-H), 1735 (C=O), 1590 (C=C) cm-1

; 1H NMR

(CDCl3, δ ppm): 7.85 (1H, d, J=3.7, H-8), 7.3-7.4 (2H, m, H-6, H-7), 6.97 (1H, d, J=3.4,

H-5), 3.47 (2H, s, H-4); 13

C NMR (CDCl3, δ ppm): 165.5 (C3), 147.1 (C1), 137.2 (C4a),

134.4 (C6), 131.5 (C8a), 130.7 (C8), 129.7 (C5), 127.5 (C7), 38.2 (C4); MS (70eV): m/z

(%) ; 162 [M+] (25), 134 (43), 118 (100), 90 (32); Anal. Calcd. for C9H6O3: C, 66.66 H,

3.70; Found: C, 66.53 H, 3.59.

General procedure for 3-alkyl/arylisocoumarins (4a-j)

A mixture of aliphatic/aromatic carboxylic acids (2a-j) (1 mmol) and thionyl

chloride (1.2 mmol) was refluxed for 1 hr in the presence of a drop of DMF. The

completion of reaction was determined by stoppage of evolution of gas. Excess of the

thionyl chloride was rotary evaporated to afford acid chlorides (3a-j).

A solution of homophthalic anhydride (1) (2.00 mmol) in acetonitril (12mL) was

added to a solution of N,N,N’,N’-tetramethylguanidine (TMG) (2.20 mmol) in acetonitril

(5mL) over 36 min maintaining an internal temperature of 0°C. Triethylmine (4.0 mmol)

was added in one portion. Acid chlorides (3a-j) (3.20 mmol) were added over 3 min and

the mixture was stirred for an additional 18 min. After the completion of reaction the

128

cooling bath was removed and reaction was allowed to warm to room temperature. The

reaction mixture was quenched by the addition of HCl (1M, 5mL). The two phases were

separated and organic layer was washed with saturated sodium chloride solution and then

dried (Na2SO4) prior to removal of solvent under reduced pressure to dryness.

Isocoumarins (4a-j) were then purified by preparative thin layer chromatography using

(petroleum ether and ethyl acetate, 7:3) as eluant.

3-(3-Fluorophenyl)isocoumarin (4a)

Yield 79%; m. p. 91-92 °C; Rf. 0.8; IR (KBr): 2980 (C-H), 1730 (C=O), 1615 (C=C) cm-

1;

1H NMR (CDCl3, δ ppm) 8.14 (1H, s, H-2’), 7.56-7.70 (2H, m, H-4’, H-5’), 7.4 (1H, d,

J=2.1, H-6’), 7.3 (2H, d, J=7.8, H-5, H-8), 7.22 (1H, dd, J=1.8, 2.1, H-6), 7.15 (1H, dd,

J=2-4, 2-4, H-7), 6.87 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 164.5 (C1), 159.2 (C3’),

143.2 (C3), 138.4 (C4a), 134.7 (C6), 132.2 (C1’), 130.5 (C8), 129.4 (C5’), 128.1 (C8a),

127.6 (C7), 126.2 (C5), 122.1 (C6’), 116.2 (C4’), 112.5 (C2’), 104.2 (C4); MS (70eV):

m/z (%) 240 [M+] (100), 212 (45), 145 (53), 117 (27); Anal. Calcd. for C15H9FO2: C,

75.00 H, 3.75 Found: C, 74.93 H, 3.69.

3-(4-Fluorophenyl)isocoumarin (4b)

Yield 70%; m. p. 109-110 °C; Rf. 0.6; IR (KBr): 3020 (C-H), 1725 (C=O), 1590 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm) 8.70 (2H, d, J=7.8, H-3’, H-5’), 7.77 (2H, d, J=3, H-2’,

H-6’), 7.72 (1H, d, J=1.2, H-5), 7.51 (3H, m, H-6, H-7, H-8), 6.95 (1H, s, H-4); 13

C NMR

(CDCl3, δ ppm) 168.2 (C1), 158.7 (C4’), 141.7 (C3), 136.3 (C4a), 133.5 (C6), 131.9

(C8), 129.5 (C8a), 128.2 (C2’, C6’), 127.6 (C7), 126.3 (C5), 125.7 (C1’), 119.6 (C3’,

C5’), 106.4 (C4); MS (70eV): m/z (%) 240 [M+] (100), 212 (45), 145 (53), 117 (27);

Anal. Calcd. for C15H9FO2: C, 75.00 H, 3.75 Found: C, 74.93 H, 3.69.

3-(2-Chlorobenzyl)isocoumarin (4c)

Yield 87%; m. p. 154-156 °C; Rf. 0.5; IR (KBr): 2860 (C-H), 1738 (C=O), 1558 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm) 7.48-7.51 (2H, m, H-6, H-7), 7.58 (1H, d, J=1.6, H-5),

7.65 (1H, d, J=1.5, H-8), 7.81 (1H, dd, J=1.5, H-5’), 7.85 (1H, d, J=1.5, H-6’), 8.1 (1H,

dd, J=1.5, 1.8, H-4’), 8.9 (1H, d, J=8.1), 2.0 (2H, s, CH2), 6.52 (1H, s, H-4); 13

C NMR

(CDCl3, δ ppm) 165.4 (C1), 143.7 (C3), 138.4 (C4a), 137.5 (C1’), 135.6 (C2’), 133.2

(C6), 131.6 (C8), 130.4 (C6’), 129.1 (C8a), 128.5 (C3’), 127.4 (C7), 126.1 (C4’), 125.5

(C5’), 123.3 (C5), 106.7 (C4), 35.2 (CH2); MS (70eV): m/z (%) 270 [M+] (100), 272

129

[M+2] (70), 242 (49), 117 (23); Anal. Calcd. for C16H11ClO2: C, 70.97 H, 4.06 Found: C,

70.89 H, 3.97.

3-(2-Bromophenyl)isocoumarin (4d)

Yield 75%; m. p. 119-120 °C; Rf. 0.8; IR (KBr): 3025 (C-H), 1710 (C=O), 1590 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm) 7.81 (1H, d, J= 2.4, H-3

’), 7.55-7.65 (3H, m, H-4

’,H-5

’,

H-6’), 7.2-7.3 (4H, m, H-5, H-6, H-7, H-8), 6.75 (1H, s, H-4);

13C NMR (CDCl3, δ ppm)

158.7 (C1), 141.1 (C3), 136.8 (C4a), 135.5 (C1’), 134.6 (C6), 131.6 (C3’), 130.7 (C4’),

129.6 (C8), 128.5 (C8), 127.6 (C6’), 126.3 (C7), 125.5 (C5’), 124.2 (C5), 118.6 (C2’),

102.7 (C4); MS (70eV): m/z (%) 300 [M+] (100), 302 [M+2] (98), 272 (43), 226 (40),

145 (60), 117 (13); Anal. calcd. for C15H9BrO2: C, 59.80 H, 2.99 Found: C, 59.73 H,

2.91.

3-(3-Iodophenyl)isocoumarin (4e)

Yield 75%; m. p. 107-108 °C; Rf. 0.7; IR (KBr): 3010 (C-H), 1725 (C=O), 1580 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm) 8.15 (1H, s, H-2’), 7.96 (1H, d, J=9, H-4’), 7.67 (1H, d,

J=8.1, H-6’), 7.61 (1H, dd, J=4.8, 3.3, H-5’), 7.51-7.55 (4H, m, H-5, H-6, H-7, H-8), 6.96

(1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 163.7 (C1), 145.6 (C3), 139.2 (C4a), 136.2 (C4’),

135.7 (C6), 134.5 (C2’), 133.1 (C1’), 130.8 (C8), 129.4 (C5’), 128.6 (C8a), 127.3 (C7),

126.8 (C5), 125.3 (C6’), 112.5 (C3’), 104.2 (C4); MS (70eV): m/z (%) 348 [M+] (100),

320 (54), 145 (64), 117 (19); Anal. calcd. for C15H9IO2: C, 51.72 H, 2.58 Found: C, 51.64

H, 2.51.

3-(2,4-Dichlorophenyl)isocoumarin (4f)

Yield 89%; m. p. 182-183 °C; Rf. 0.6; IR (KBr): 2970 (C-H), 1705 (C=O), 1620 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm) 7.6 (1H, d, J=0.9, H-3’), 7.5 (1H, d, J=13.2, H-5’), 7.4

(1H, d, J=8.5,H-6’), 7.1-7.35 (4H, m, H-5, H-6, H-7, H-8), 6.95 (1H, s, H-4); 13

C NMR

(CDCl3, δ ppm) 164.6 (C1), 141.5 (C3), 138.6 (C4a), 136.5 (C4’), 135.6 (C2’), 134.2

(C6), 132.8 (C8), 131.5 (C3’), 130.9 (C1’), 128.5 (C8a), 127.8 (C6’), 126.5 (C7), 125.7

(C5’), 123.6 (C5), 103.7 (C4); MS (70eV): m/z (%) 291 [M+] (100), 293 [M+2] (70), 295

[M+4] (13), 263 (49), 117 (23); Anal. calcd. for C15H8Cl2O2: C, 61.85 H, 2.74 Found: C,

61.77 H, 2.67.

130

3-(2-Chloro-4-fluorophenyl)isocoumarin (4g)

Yield 84%; m. p. 176-177 °C; Rf. 0.7; IR (KBr): 2990 (C-H), 1705 (C=O), 1595 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm) 7.80 (1H, s, H-3

’), 7.78 (1H, d, J=2.7 H-5

’), 7.19 (1H, d,

J=2.4, H-6’), 7.15 (4H, m, H-5, H-6, H-7, H-8), 6.77 (1H, s, H-4);

13C NMR (CDCl3, δ

ppm) 167.3 (C1), 159.5 (C4’), 143.6 (C3), 137.5 (C4a), 135.4 (C2’), 134.5 (C6), 132.1

(C8), 131.0 (C8a), 130.4 (C6’), 129.4 (C1’), 127.3 (C7), 126.2 (C5), 121.3 (C3’), 117.4

(C5’), 105.3 (C4); MS (70eV): m/z (%) 274.5 [M+] (100), 276.5 (70), 246.5 (44), 145

(62), 117 (19); Anal. calcd. for C15H8ClFO2: C, 65.57 H, 2.91 Found: C, 65.49 H, 2.84.

3-(3-Nitrophenyl)isocoumarin (4h)

Yield 81%; m. p. 109 °C; Rf. 0.5; IR (KBr): 2893 (C-H), 1734 (C=O), 1512 (C=C) cm-1

;

1H NMR (CDCl3, δ ppm) 8.4 (1H, s, H-2’), 8.2 (1H, d, J=8.2,H-4’), 7.7-7.5 (2H, m, H-5’,

H-6’), 7.2-7.4 (4H, m, H-5, H-6, H-7, H-8), 7.1 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm)

162.3 (C1), 149.3 (C3’), 141.4 (C3), 137.5 (C4a), 135.1 (C6), 132.5 (C1’), 131.6 (C6’),

130.5 (C5’), 129.1 (C8), 128.4 (C8a), 127.3 (C7), 126.1 (C5), 123.5 (C2’), 121.8 (C4’),

105.1 (C4); MS (70eV): m/z (%) 267 [M+] (100), 239 (59), 145 (61), 117 (21); Anal.

calcd. for C15H9NO4: C, 67.41 H, 3.37 N, 5.24 Found: C, 67.33 H, 3.31 N, 5.15.

3-(2-Chloropyridyl)isocoumarin (4i)

Yield 85%; m. p. 122-124 °C; Rf. 0.6; IR (KBr): 2882 (C-H), 1743 (C=O), 1543 (C=C)

cm-1

; 1H NMR (CDCl3, δ ppm) 8.7 (1H, d, J=1.6, H-3’), 8.5 (1H, d, J=2.1, H-5’), 8.0

(1H, dd, J=2.2, 2.4, H-4’), 7.6 (1H, d, J=1.5, H-8), 7.2-7.5 (3H, m, H-5, H-6, H-7), 6.95

(1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 168.2 (C1), 157.8 (C1’), 148.6 (C3’), 146.3 (C3),

140.8 (C5’), 138.6 (C4a), 134.2 (C6), 131.5 (C8), 129.5 (C8a), 127.9 (C7), 126.3 (C5),

124.1 (C4’), 121.6 (C6’), 104.3 (C4); MS (70eV): m/z (%) 257.5 [M+], (100), 259.5

[M+2] (70), 229.5 (34), 145 (63), 117 (17); Anal. calcd. for C14H8ClNO2: C, 65.24 H,

3.10 N, 5.43 Found: C, 65.17 H, 3.04 N, 5.37.

3-Pentadecylisocoumarin (4j)

Yield 76%; m. p. 71-73 °C; Rf. 0.55; IR (film): 2918 (C-H), 2849 (Sp3 C-H), 1728

(C=O), 1604 (C=C), cm-1

; 1H NMR (CDCl3, δ ppm) 8.25 (1H, d, J = 8.16, H-8), 7.65

(1H, m, H-6), 7.49 (1H, td, J = 0.88, 7.28, H-7), 7.34 (1H, d, J = 8.16, H-5), 6.24 (1H, s,

H-4), 2.52 (2H, t, J = 7.08, 2H, H-1’), 1.70 (2H, p, J = 8.4, H2’), 1.28 (24H, brs, H3’-

H14’), 0.87 (3H, t, J = 6.28, H-15’); 13

C NMR (CDCl3, δ ppm): 165.4 (C1), 152.3 (C3),

131

138.5 (C4a), 134.3 (C6), 131.5 (C8), 129.4 (C8a), 128.7 (C7), 126.2 (C5), 105.3 (C4),

33.9 (C1’), 24.2 (C2’), 29.8 (C3’), 29.6 (C4’), 29.4 (C5’), 28.8 (C6’), 28.6 (C7’), 28.2

(C8’), 27.9 (C9’), 27.6 (C10’), 26.8 (C11’), 25.4 (C12’), 24.6 (C13’), 14.3 (C14’), 11.7

(C15’); MS (70eV): m/z (%) 356 [M+] (100), 328 (34), 145 (37), 117 (21); Anal. calcd.

for C24H36O2: C, 80.89 H, 10.11 Found: C, 80.81 H, 10.04.

General procedure for the conversion of isocoumarins into 1(2H)-

isoquinolones (5a–j)

A mixture of isocoumarins (4a-j) (10 mmol) and formamide (10 mmol) was

refluxed for 2-4 h. On completion of the reaction, followed by TLC, the solution was

poured into water (300 ml). The resulting precipitates were filtered and recrystallized

from ethyl acetate to afford 1(2H)-isoquinolones (5a–j).

3-(3-Flourophenyl) isoquinolin-1(2H)-one (5a)

Yield 84%; m. p. 216-218 °C; Rf. 0.65; IR (KBr): 3332 (NH), 2825 (C-H), 1656 (C=O),

1517 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 10.3 (1H, s, NH), 8.4 (1H, d, J=1.2, H-4’),

7.72 (1H, d, J=0.9, H-2’), 7.70 (1H, d, J=1.5, H-6’), 7.54 (1H, dd, J=1.8, 1.6, H-5’), 7.23

(1H, d, J=2.4, H-8), 7.1-7.2 (3H, m, H-5, H-6, H-7), 6.83 (1H, s, H-4); 13

C NMR (CDCl3,

δ ppm) 168.2 (C1), 156.2 (C3’), 139.4 (C3), 137.5 (C4a), 135.4 (C6), 131.7 (C1’), 130.8

(C8), 129.9 (C5’), 128.5 (C8a), 127.2 (C7), 126.7 (C5), 123.4 (C6’), 117.4 (C4’), 113.7

(C2’), 103.6 (C4); MS (70eV): m/z (%) 239 [M+] (100), 211 (41), 144 (47), 117 (19);

Anal. calcd. for C15H10FNO: C, 75.31 H, 4.18 N 5.85 Found: C, 75.24 H, 4.11 N, 5.78.

3-(4-Flourophenyl)isoquinolin-1(2H)-one (5b)

Yield 82%; m. p. 222-224 °C; Rf. 0.55; IR (KBr): 3320 (NH), 2870 (C-H), 1630 (C=O),

1527 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 10.54 (1H, s, NH), 8.4 (2H, d, J=8.4, H-3’,

H-5’), 7.7 (2H, d, J=1.8, H-2’,H-6’), 7.63 (1H, d, J=1.2, H-8), 7.5-7.6 (2H, m, H-6, H-7),

7.4 (1H, dd, J=1.2, 1.1, H-5), 6.8 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 166.5 (C1),

159.4 (C4’), 142.3 (C3), 137.4 (C4a), 134.2 (C6), 131.6 (C8), 129.8 (C8a), 128.5 (C2’,

C6’), 127.1 (C7), 126.6 (C5), 125.2 (C1’), 119.2 (C3’, C5’), 105.4 (C4); MS (70eV): m/z

(%) 239 [M+] (100), 211 (41), 144 (47), 117 (19); Anal. calcd. for C15H10FNO: C, 75.31

H, 4.18 N 5.85 Found: C, 75.24 H, 4.11 N, 5.78.

132

3-(2-Chlorobenzyl)isoquinolin-1(2H)-one (5c)

Yield 85%; m. p. 188-190 °C; Rf. 0.6; IR (KBr): 3348 (NH), 2860 (C-H), 1638 (C=O),

1558 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 9.53 (1H, s, NH), 9.1 (1H, d, J=8.1, H-3’),

8.3 (1H, dd, J=1.5, 1.8, H-4’), 7.89 (1H, d, J=1.5, H-6’), 7.85 (1H, dd, J=1.5, 1.6, H-5’),

7.7 (1H, d, J=1.5, H-8), 7.6 (1H, d, J=1.5,H-5), 7.5-7.55 (2H, m, H-6,H-7), 6.66 (1H,

s,H-4), 2.19 (2H, s, CH2); 13

C NMR (CDCl3, δ ppm) 167.5 (C1), 141.3 (C3), 137.5 (C4a),

136.4 (C1’), 135.2 (C2’), 134.5 (C6), 132.8 (C8), 131.3 (C6’), 129.7 (C8a), 128.8 (C3’),

127.5 (C7), 126.5 (C4’), 124.9 (C5’), 122.8 (C5), 104.6 (C4), 37.6 (CH2); MS (70eV):

m/z (%) 269 [M+] (100), 271 [M+2] (70), 241 (37), 117 (23); Anal. calcd. for

C16H12ClNO: C, 71.24 H, 4.45 N, 5.19 Found: C, 71.19 H, 4.37 N, 5.13.

3-(2-Bromophenyl)isoquinolin-1(2H)-one (5d):

Yield 82%; m. p. 170-172 °C; Rf. 0.65; IR (KBr): 3341 (NH), 2864 (C-H), 1635 (C=O),

1535 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 9.4 (1H, s, NH), 9.12 (1H, d, J=8.4, H-3’),

8.38 (1H, d, J=1.5, H-6’), 7.66-7.75 (2H, m, H-4’,H-5’), 7.63 (1H, d, J=2.7, H-8), 7.45-

7.55 (2H, m, H-6, H-7), 7.34 (1H, d, J=1.8, H-5), 6.63 (1H, s, H-4); 13

C NMR (CDCl3, δ

ppm) 163.6 (C1), 143.1 (C3), 137.3 (C4a), 134.6 (C1’), 133.8 (C6), 132.3 (C3’), 130.3

(C4’), 129.5 (C8), 128.4 (C8), 127.6 (C6’), 126.7 (C7), 125.2 (C5’), 123.9 (C5), 117.3

(C2’), 104.5 (C4); MS (70eV): m/z (%) 299 [M+] (100), 301 [M+2] (98), 271 (43), 144

(60), 117 (19); Anal. calcd. for C15H10BrNO: C, 60.00 H, 3.33 N, 4.66 Found: C, 59.93

H, 3.26 N, 4.61.

3-(3-Iodophenyl)isoquinolin-1(2H)-one (5e):

Yield 84%; m. p. 226-228 °C; Rf. 0.7; IR (KBr): 3353 (NH), 2892 (C-H), 1643 (C=O),

1533 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 10.6 (1H, s ,NH), 8.5 (1H, s, H-2’), 8.2 (1H,

d, J=13.8, H-4’), 8.1 (1H, t, J=1.8, H-5’), 7.7 (1H, d, J=1.2, H-6’), 7.6 (1H, d, J=1.2, H-

8), 7.5 (1H, d, J=1.2, H-5), 7.5-7.54 (2H, m, H-6, H-7), 6.8 (1H, s, H-4); 13

C NMR

(CDCl3, δ ppm) 166.3 (C1), 143.5 (C3), 138.3 (C4a), 136.7 (C4’), 135.3 (C6), 134.5

(C2’), 133.4 (C1’), 131.7 (C8), 129.7 (C5’), 128.2 (C8a), 127.5 (C7), 126.5 (C5), 125.7

(C6’), 113.6 (C3’), 105.3 (C4); MS (70eV): m/z (%) 347 [M+] (100), 319 (47), 144 (34),

117 (13); Anal. calcd. for C15H10INO: C, 51.87 H, 2.88 N, 4.03 Found: C, 51.81 H, 2.82

N, 3.96.

133

3-(2,4-Dichlorophenyl)isoquinolin-1(2H)-one (5f):

Yield 78%; m. p. 221-222 °C; Rf. 0.6; IR (KBr): 3330 (NH), 2862 (C-H), 1650 (C=O),

1550 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 10.9 (1H, s, NH), 9.0 (1H, d, J=8.1, H-5’),

8.4 (1H, d, J=7.8, H-6’), 7.88 (1H, s, H-3’), 7.7 (1H , d, J=6.9, H-8), 7.61-7.67 (2H, m

,H-6, H-7), 7.5 (1H, d, J= 5.7, H-5), 6.6 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 167.3

(C1), 142.4 (C3), 137.3 (C4a), 136.1 (C4’), 135.2 (C2’), 134.5 (C6), 133.2 (C8), 132.9

(C3’), 131.2 (C1’), 129.8 (C8a), 128.7 (C6’), 126.2 (C7), 125.4 (C5’), 124.1 (C5), 104.2

(C4); MS (70eV): m/z (%) 290 [M+] (100), 292 [M+2] (70), 294 [M+4] (13), 262 (38),

117 (27); Anal. calcd. for C15H9Cl2NO: C, 60.06 H, 3.10 N, 4.82 Found: C, 59.98 H,

3.03 N, 4.74.

3-(2-Chloro-4-Flourophenyl)isoquinolin-1(2H)-one (5g):

Yield 80%; m. p. 213 °C; Rf. 0.55; IR (KBr): 3381 (NH), 2876 (C-H), 1655 (C=O), 1550

(C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 10.0 (1H, s, NH), 8.4 (1H, d, J=8.1, H-5’), 8.2

(1H, s, H-3’), 7.9 (1H, d, J=1.2, H-6’), 7.8 (1H, d, J=1.2, H-8), 7.7 (1H, d, J =1.2, H-5),

7.5-7.6 (2H, m, H-6, H-7), 6.65 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 168.4 (C1), 158.4

(C4’), 141.5 (C3), 138.2 (C4a), 136.2 (C2’), 135.1 (C6), 132.3 (C8), 131.5 (C8a), 130.8

(C6’), 129.6 (C1’), 128.2 (C7), 127.8 (C5), 124.2 (C3’), 118.8 (C5’), 104.8 (C4); MS

(70eV): m/z (%) 273.5 [M+] (100), 275.5 (70), 245.5 (56), 144 (61), 117 (21); Anal.

calcd. for C15H9ClFNO: C, 65.81 H, 3.29 N, 5.11 Found: C, 65.75 H, 3.22 N, 5.04.

3-(3-Nitrophenyl)isoquinolin-1(2H)-one (5h):

Yield 84%; m. p. 230-232 °C; Rf. 0.7; IR (KBr): 3339 (NH), 2821 (C-H), 1634 (C=O),

1512 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 8.75 (1H, s, NH), 8.6 (1H, s, H-2’), 8.4 (1H,

d, J=8.4, H-4’), 7.7-7.2 (2H, m, H-5’, H-6’), 7.4-7.6 (4H, m, H-5, H-6, H-7, H-8), 6.9

(1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 164.7 (C1), 151.5 (C3’), 143.5 (C3), 138.2 (C4a),

136.2 (C6), 132.9 (C1’), 131.8 (C6’), 130.7 (C5’), 129.5 (C8), 128.6 (C8a), 127.7 (C7),

126.2 (C5), 124.1 (C2’), 122.3 (C4’), 104.2 (C4); MS (70eV): m/z (%) 266 [M+] (100),

238 (46), 144 (51), 117 (14); Anal. calcd. for C15H10N2O3: C, 67.66 H, 3.75 N, 10.52

Found: C, 67.59 H, 3.69 N, 19.45.

3-(2-Chloropyridyl)isoquinolin-1(2H)-one (5i):

Yield 82%; m. p. 210-212 °C; Rf. 0.6; IR (KBr): 3353 (NH), 2882 (C-H), 1663 (C=O),

1543 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 10.2 (1H, s, NH), 8.8 (1H, d, J=1.8, H-3’),

134

8.7 (1H, d, J=2.2, H-5’), 8.1 (1H, dd, J=2.4, 2.3, H-4’), 7.8 (1H, d, J=1.5, H-8), 7.5-7.7

(3H, m, H-5, H-6, H-7), 6.84 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm) 169.5 (C1), 159.1

(C1’), 149.5 (C3’), 144.5 (C3), 141.5 (C5’), 138.2 (C4a), 134.3 (C6), 132.1 (C8), 129.8

(C8a), 128.2 (C7), 127.1 (C5), 125.2 (C4’), 122.4 (C6’), 105.7 (C4); MS (70eV): m/z (%)

256.5 [M+], (100), 258.5 [M+2] (70), 228.5 (31), 144 (58), 117 (26); Anal. calcd. for

C14H9ClN2O: C, 65.49 H, 3.50 N, 10.91 Found: C, 65.42 H, 3.43 N, 10.84.

3-Pentadecylisoquinolin-1(2H)-one (5j):

Yield 76%; m. p. 71-73 °C; Rf. 0.55; IR (KBr): 3323 (NH), 2876 (C-H), 1652 (C=O),

1513 (C=C) cm-1

; 1H NMR (CDCl3, δ ppm) 9.62 (1H, s, NH), 8.2 (1H, d, J=13.5, H-8),

7.7-7.74 (2H, m, H-6, H-7), 7.6 (1H, d, J=1.2, H-5), 6.9 (1H, s, H-4), 2.3 (2H, t, J=7.5,

H-1’), 1.5-1.7 (26H, m, H-2’-H-14’), 0.9 (3H, t, J=5.1, H-15’); 13

C NMR (CDCl3, δ

ppm): 167.5 (C1), 151.7 (C3), 137.9 (C4a), 135.1 (C6), 132.2 (C8), 129.7 (C8a), 128.3

(C7), 126.3 (C5), 104.5 (C4), 33.2 (C1’), 32.3 (C2’), 29.7 (C3’), 29.5 (C4’), 29.3 (C5’),

28.6 (C6’), 28.1 (C7’), 27.9 (C8’), 27.6 (C9’), 27.1 (C10’), 26.8 (C11’), 25.4 (C12’), 24.6

(C13’), 14.3 (C14’), 11.7 (C15’); MS (70eV): m/z (%) 355 [M+] (100), 327 (39), 144

(49), 117 (18); Anal. calcd. for C24H37NO: C, 81.12 H, 10.42 N, 3.94 Found: C, 81.05

H, 10.35 N, 3.88.

7.3 RESULTS AND DISCUSSION

Homophthalic acid was converted into corresponding anhydride (1) by refluxing

homophthalic acid with acetic anhydride in the presence of dry toluene as solvent. The

formation of the anhydride (1) was confirmed by the presence of absorption at 1735cm-1

and disappearance of the absorption for hydroxyl groups in IR spectrum.

O

O

OHOH O

O

O

(1)

Acetic Anhydride

Dry Toluene

The substituted aromatic carboxylic acids (2a-j) were converted into their

respective acid chlorides by reacting them with thionyl chlorides in the presence of

catalytic amount of DMF. Acid chlorides (3a-j) were then condensed with homophthalic

anhydride (1) in the presence of triethyl amine and tetramethyl guanidine to afforded 3-

135

aryl isocoumarins (4a-j). These isocoumarins were purified by preparative thin layer

chromatography using petroleum ether and ethyl acetate, 3:1 as eluant.

The synthesized isocoumarins (4a-j) showed the characteristic absorption for

lactonic carbonyl at 1713-1736 cm-1

. The detailed physical and FTIR spectral data of the

isocoumarins (4a-j) are shown in Table 7.1. In 1H NMR, spectrum a characteristic singlet

appeared for H-4 at δ 6.1-6.35 ppm and the lactonic carbonyl carbon in 13

C NMR showed

the peak at δ 158-170 ppm.

R

O

OH

+ SOCl2 R

O

Cl

(3a-j)(2a-j)

R is same as in (2a-j)

R

O

Cl

+O

O

O

(1)

O

O

R

(4a-j)(3a-j)

TMG / (C2H5)3N

CH3CN

R is same as in (2a-j)(2a-j ) R=

2a=3-F-C6H4 ; 2b=4-FC6H42c= 2-CIC6H4CH2; 2d=2-BrC6H42e=3-I-C6H4; 2f=2,4-DiClC6H3122g=2-F-4-ClC6H3; 2h=4-NO2C6H42i=2-ClC6H3N; 2j=C15H31

Scheme 7.1 Synthesis of 3-phenyl substituted isocoumarins (4a-j)

Table 7.1 Physical constants and FTIR spectral data of isocoumarins (4a-j)

Compds. M. P.

(°C) Rf

Yield

(%) Ar-C-H C-H C=O C=C

4a 107-108 0.7 72 3011 - 1723 1583

4b 119-120 0.8 75 3021 - 1715 1615

4c 91-92 0.8 79 3005 - 1719 1597

4d 109-110 0.6 70 3010 - 1725 1590

4e 109 0.5 81 3019 - 1716 1612

4f 182-183 0.6 89 3008 - 1735 1599

4g 176-177 0.7 84 3012 - 1727 1583

4h 154-156 0.5 87 3010 2882 1731 1618

4i 122-124 0.6 85 3017 - 1720 1593

4j 71-73 0.8 76 3012 2872 1725 1588

Pet. Ether: Ethyl Acetate (3:1)

136

An equimolar mixture of the isocoumarin (4a-j) and methanamide was refluxed

for 2-4 hours to afford corresponding 1(2H)-isoquinolones (5a-j). The products were

obtained in 76-85 % yields in high purity. The progress of the reaction was followed by

TLC. The successful substitution was initially indicated by appearance of a fluorescent

blue spot under longer wave length of UV lamp, having Rf values lower than that of the

parent isocoumarin.

The products were further characterized by comparison of their m. p., IR, 1H,

13C

NMR and mass spectral data with those of the corresponding isocoumarins. Thus, a shift

of lactonic carbonyl absorption from 1710-1730 cm-1

to 1630-1650 cm-1

and appearance

of absorption at 3220-3380 cm-1

for NH was noted in the IR spectra. The physical

constants and FTIR spectral data of the compounds (5a-j) is shown in table 7.2. In the 1H

NMR a downfield shift of the characteristic H-4 proton of the isocoumarins at δ 6.0-6.2

to δ 6.6-6.9 ppm in isoquinolones was observed besides, appearance of NH absorption at

δ 9.4-10.8 ppm shown in table 7.3. A variety of substituents on the aryl ring are well-

tolerated, and the reaction leads to completion in all the cases. The generality of the

conversion was indicated by substrates bearing an aralkyl group (5c), heterocyclyl (5i) or

a long aliphatic chain (5j) at C-3 position.

O

O

R

NH

O

R

2-4h; 76-85 %

(4a-j ) R=

4a=3-F-C6H4 ; 4b=4-FC6H44c= 2-CIC6H4CH2; 4d=2-BrC6H44e=3-I-C6H4; 4f=2,4-DiClC6H3124g=2-F-4-ClC6H3; 4h=4-NO2C6H44i=2-ClC6H3N; 4j=C15H31

(5a-j)

R= same as in 4a-j

OH2N

Scheme 7.2 Synthesis of 3-phenyl substituted isoquinolones (5a-j)

137

Table 7.2 Physical constants and FTIR spectral data of isoquinolones (5a-j)

Compds. M. P.

(°C) Rf

Yield

(%)

Ar-C-

H C-H C=O C=C N-H

5a 226-228 0.7 84 3013 - 1643 1533 3353

5b 170-172 0.8 82 3023 - 1635 1535 3341

5c 216-218 0.8 84 3015 - 1656 1517 3332

5d 222-224 0.6 82 3011 - 1630 1527 3320

5e 230-232 0.5 84 3009 - 1634 1512 3339

5f 221-222 0.6 78 3010 - 1651 1550 3330

5g 213-214 0.7 80 3013 - 1655 1543 3381

5h 188-190 0.5 85 3010 2860 1638 1558 3348

5i 210-212 0.6 82 3018 - 1663 1543 3353

5j 71-73 0.8 76 3001 2872 1652 1523 3323

Pet. Ether: Ethyl Acetate (3:1)

Table 7.3 Comparison of the δ values of H-4 among isocoumarins and isoquinolones

Entry Compd. R

1H NMR δ (ppm)

H-4 (4) H-4 (5) N-H (5)

1 5a 3-FC6H4 6.1 6.83 10.25

2 5b 4-FC6H4 6.4 6.80 10.54

3 5c 2-ClCH2C6H4 6.24 6.66 9.53

4 5d 2-BrC6H4 6.2 6.63 9.4

5 5e 3-IC6H4 6.34 6.8 9.8

6 5f 2,4-DiClC6H3 6.21 6.66 10.89

7 5g 2-Cl-4-FC6H3 6.35 6.65 10.05

8 5h 4-NO2C6H4 6.26 6.94 8.79

9 5j 2-Cl-C6H3N 6.3 6.84 10.2

10 5k C15H31 6.24 6.89 6.62

138

Synthesis of 3-phenyl substituted-1H-isochromen-1-thiones

7.4 EXPERIMENTAL

Melting points were recorded using a digital Gallenkamp (SANYO) model MPD

BM 3.5 apparatus and are uncorrected. 1H NMR and the

13C NMR spectra were

determined as CDCl3 solutions at 300 MHz and 100 MHz respectively, on a Bruker AM-

300 machine. FT IR spectra were recorded using an FTS 3000 MX spectrophotometer;

Mass Spectra (EI, 70eV) on a GCMS instrument and elemental analyses with a LECO-

183 CHNS analyzer. The reactions were carried out in an unmodified domestic

microwave oven (MW 900 W, frequency 2450 MHz, Power level 1, Dawlance,

Pakistan). The analytical TLC was carried out using recoated plated from Merck and

thick layer chromatography using silica gel from Merck.

General procedure for the conversion of isocoumarins into 1-1H-

isochromene-1-thiones (2a–j)

A homogenized mixture of isocoumarin (6a-j) (1 mmol) and Lawesson’s reagent

(0.5-0.6 mmol) was irradiated for 1-3 min in an alumina bath inside the microwave oven.

The progress of the reaction was followed by TLC examination using hexane/ethyl

acetate (9:1). On completion, the reaction mixture was diluted with ethyl acetate and

subjected to thick layer chromatography using same solvent system. Elution using ethyl

acetate followed by concentration afforded the products (7a–j) which crystallized on

standing as yellow needles or plates.

3-(3-Fluorophenyl)-1H-isochromene-1-thione (7a)

Yield: 78%; m. p. 109-113 °C; Rf 0.8; IR (KBr): 2980 (C-H), 1615 (C=C), 1190 (C=S)

cm-1

. 1H NMR (CDCl3, δ ppm): 8.34 (1H, s, H-2’), 7.66-7.75 (2H, m, H-4’, H-5’), 7.62

(1H, d, J=2.1, H-6’), 7.53 (2H, d, J=7.8, H-5, H-8), 7.44 (1H, dd, J=1.8, 2.1, H- 6), 7.15

(1H, dd, J=2.4, 2.4, H-7), 6.98 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm): 203 (C1), 164

(C3), 162 (C3’), 152 (C8a), 137 (C1’), 135 (C6), 134 (C5), 130 (C2’,C4’), 129 (C8), 126

(C7), 120 (C6’,C5’), 112 (C 4a), 102 (C4); MS (70eV): m/z (%); 256 [M+] (100), 161

(67), 95 (48). Anal. calcd. For C15H9OSF: C, 70.31 H, 3.51 S, 12.50. Found. C, 70.25 H,

3.45 S, 11.45.

139

3-(4-Fluorophenyl)-1H-isochromene-1-thione (7b)

Yield: 95%; m. p. 138 °C; Rf. 0.6, IR (KBr): 3020 (C-H), 1590 (C=C), 1195 (C=S) cm-1

.

1H NMR (CDCl3, δ ppm): 8.73 (2H, d, J=7.8, H-3’, H-5’), 7.97 (2H, d, J=3, H-2’, H-6’),

7.72 (1H, d, J=1.2, H-5), 7.51 (3H, m, H-6, H-7, H-8), 7.08 (1H, s, H-4); 13

C NMR

(CDCl3, δ ppm): 200 (C-1), 165 (C-3), 162 (C-4’), 155 (C-8a), 135 (C-1’), 132 (C-3’,C-

5’), 130 (C-8,C-5), 129 (C-7), 127 (C-2’,C-6’), 116 (C-4a), 104 (C-4); MS (70eV): m/z

(%) 256 [M+] (100), 161 (52), 95 (38). Anal. calcd. For C15H9OSF: C, 70.31 H, 3.51 S,

12.50. Found. C, 70.19 H, 3.41 S, 11.41.

3-(4-Chlorophenyl)-1H-isochromene-1-thione (7c)

Yield: 89%; m. p. 128-130 °C; Rf. 0.7, IR (KBr) 3025 (C-H), 1615 (C=C), 1171 (C=S)

cm-1

; 1H NMR (CDCl3, δ ppm): 7.85 (2H, d, J=1.8, H-3’,H-5’), 7.83 (2H, d, J=2.1, H-2’-

H-6’), 7.75 (1H, d, J=1.5, H-5), 7.73 (1H, d, J=1.2, H-8), 7.40 (2H, m, H-6, H-7), 6.96

(1H, s, H-4); 13

C NMR (CDCl3, δ ppm): 195 (C-1), 162 (C-3), 152 (C-4’), 152 (C-8a),

137 (C-6), 136 (C-5), 135 (C-3’,C-5’), 130 (C-2’,C-6’), 129 (C-1’), 128 (C-8), 126 (C-

7), 120 (C-4a), 102 (C-4); MS (70eV): m/z (%) 272.5 [M+] (100), 161 (67), 111.5 (48).

Anal. calcd. For C15H9OSCl: C, 66.05 H, 3.30 S, 11.74. Found. C, 65.76 H, 3.22 S,

11.66.

3-(2-Bromophenyl)-1H-isochromene-1-thione (7d)

Yield: 81%; m. p. Oil; Rf. 0.6, IR (KBr): 3025 (C-H), 1590 (C=C), 1079 (C=S) cm-1

; 1H

NMR (CDCl3, δ ppm): 8.03 (1H, d, J= 2.4, H-3’), 7.61-7.69 (3H, m, H-4’,5’,6’), 7.30-

7.40 (4H, m, H-5, 6, 7, 8), 6.89 (1H, s, H-4); 13

C NMR (CDCl3, δ ppm): 194 (C- 1), 164

(C-3), 152 (C-8a), 141 (C-2’), 136 (C-6), 134 (C-5), 133.8 (C-1’), 133 (C-3’), 132 (C-

4’,C-6’), 131 (C-5’), 129 (C-8), 127 (C-7), 113 (C-4a), 107 (C-4); MS (70eV): m/z (%)

316 [M+] (100), 161 (68), 155 (52). Anal. calcd. For C15H9OSBr: C, 56.96 H, 2.84 S,

10.12. Found. C, 56.87 H, 2.78 S, 10.05.

3-(3-Iodophenyl)-1H-isochromene-1-thione (7e)

Yield: 71%; m. p. 116-118 °C; Rf. 0.65; IR (KBr): 3010 (C-H), 1580 (C=C), 1085 (C=S)

cm-1

; 1H NMR (CDCl3, δ ppm): 8.25 (1H, s, H-2’), 8.06 (1H, d, J=9, H-4’), 7.77 (1H, d,

J=8.1, H-6’), 7.73 (1H, dd, J=4.8, 3.3, H-5’), 7.52-7.57 (4H, m, H-5-H-8), 6.97 (1H, s,

H-4); 13

C NMR (CDCl3, δ ppm): 197 (C-1), 158 (C-3), 155 (C-8a), 152 (C-3’), 137 (C-6),

135 (C-5), 132 (C-8), 130 (C-7), 128 (C-2’,C-4’), 109 (C-4), 125 (C- 1’), 113 (C-5’), 112

140

(C-6’), 109 (C-4a); MS (70eV): m/z (%) 364 [M+] (100), 203 (48), 161 (67). Anal. calcd.

For C15H9OSI: C, 49.45 H, 2.47 S, 8.79. Found. C, 49.37 H, 2.39 S, 8.71.

3-(2,4-Dichorophenyl)-1H-isochromene-1-thione (7f)

Yield: 74%; m. p. 123-125 °C; Rf. 0.7 IR (KBr): 2970 (C-H), 1620 (C=C), 1128 (C=S),

cm-1

; 1H NMR (CDCl3, δ ppm): 7.80 (1H, d, J=0.9, H-3’), 7.74 (1H, d, J=13.2, H-5’),

7.71 (1H, d, J=8.5, H-6’), 7.51-7.61 (4H, m, H-5-H-8), 7.03 (1H, s, H-4); 13

C NMR

(CDCl3, δ ppm): 208 (C-1), 150 (C-3), 137 (C-2’,C-4’), 136 (C-8a), 135 (C-3’), 133 (C-

1’), 131 (C-5’), 130 (C-6), 129 (C-5), 128 (C-8), 127 (C-7), 126 (C-6’), 108 (C-4a), 106

(C-4); MS (70eV): m/z (%) 307 [M+] (100), 161 (62), 146 (52). Anal. calcd. For

C15H9OSCl2: C, 58.63 H, 2.60 S, 10.42. Found. C, 58.55 H, 2.52 S, 10.37.

3-(2-Chloro-4-fluorophenyl)-1H-isochromene-1-thione (7g)

Yield: 81%; m. p. 129 °C; Rf. 0.6; IR (KBr): 2990 (C-H), 1595 (C=C), 1275 (C=S) cm-1

;

1H NMR (CDCl3, δ ppm): 7.83 (1H, s, H-3’), 7.81 (1H, d, J=2.7 H-5’), 7.29 (1H, d,

J=2.4, H-6’), 7.28 (4H, m, H-5-8), 6.97 (1H, s, H-4). 13

C NMR (CDCl3, δ ppm): 200

(C-1), 164 (C-3), 161 (C-4’), 153 (C-2’), 153 (C-8a), 135 (C-6), 134 (C-5), 133 (C-3’),

132 (C-5’), 131 (C-1’), 130 (C-6’), 129 (C-8), 127 (C-7), 114 (C-4a), 107 (C-4), MS

(70eV): m/z (%) 290.5 [M+] (100), 161 (75), 129.5 (34); Anal. calcd. For C15H9OSClF:

C, 61.96 H, 2.75 S, 11.01; Found. C, 61.85 H, 2.68 S, 10.96.

3-(4-Methoxyphenyl)-1H-isochromene-1-thione (7h)

Yield: 91%; m. p. 109-111 °C; Rf. 0.5; IR (KBr): 3015 (C-H), 1575 (C=C), 1205 (C=S)

cm-1

; 1H NMR (CDCl3, δ ppm): 7.85 (2H, d, J=2.1, H-3’, H-5’), 7.83 (2H, d, J=2.1, H-

2’-H-6’), 7.74 (1H, d, J=1.5, H-5), 7.69 (1H, d, J=1.5, H-8), 7.40-7.50 (2H, m, H-6, H-7),

6.86 (1H, s, H-4), 3.88 (3H, s, H-4’-OCH3); 13

C NMR (CDCl3, δ ppm): 203 (C-1),

162 (C-3), 161 (C-4’), 153 (C-8a), 137 (C-6), 134 (C-5), 130 (C-3’,C-5’), 129 (C-1’), 128

(C-8), 127 (C-7), 126 (C-2’,C-6’), 114 (C-4a), 100 (C-4), 55.0 (C-OCH3), MS (70eV):

m/z (%) 268 [M+] (100), 161 (63), 107 (72); Anal. calcd. For C16H12O2S: C, 71.64 H,

4.47 S, 11.94. Found. C, 71.57 H, 4.39 S, 11.87.

3-(4-Fluorobenzyl)-1H-isochromene-1-thione (7i)

Yield: 93%; m. p. 65-67 °C; Rf. 0.6; IR (KBr): 2960 (C-H), 1610 (C=C), 1194 (C=S) cm-

1;

1H NMR (CDCl3, δ ppm): 7.73 (2H, d, J = 3.1, H-3’, 5’), 7.71 (2H, d, J=3.3 H-2’, H-

6’), 7.30 (4H, m, H-5-H-8), 7.01 (1H, s, H-4), 3.64 (2H, s, CH2); 13

C NMR (CDCl3, δ

141

ppm): 176 (C-1), 167 (C-3), 164 (C-4’), 153 (C-8a), 136 (C-6), 135 (C-5), 134 (C-

3’,5’), 131 (C-2’,6’), 130 (C-1’), 129 (C-8), 127 (C-7), 114 (C-4a), 107 (C-4), 68 (CH2);

MS (70eV): m/z (%) 270 (M+] (100), 161 (62), 109 (37); Anal. calcd. for C16H11OSF: C,

71.11 H, 4.07 S, 11.85. Found. C, 71.05 H, 4.01 S, 11.78.

3-(Pentadecyl)-1H-isochromene-1-thione (7j)

Yield: 87%; m. p. 32-33 °C; Rf. 0.8; IR (KBr): 3010 (C-H), 1605 (C=C), 1272 (C=S); 1H

NMR (CDCl3, δ ppm): 8.26 (1H, d, J=8.1, H-8), 7.66-7.68 (2H, m, H-6, H-7), 7.36

(1H, d, J=7.8, H-5), 6.27 (1H, s, H-4), 2.50 (2H, t, J=7.5, H-1’), 1.73 (2H, p, J=6.6, H-

2’), 1.27-1.38 (24H, m, H-3’-H-14’), 0.89 (3H, t, J=5.4, H-15’). 13

C NMR (CDCl3, δ

ppm): 201 (C-1), 167 (C-3), 158 (C-8a), 137 (C-6), 132 (C-8), 129 (C-7), 128 (C-5), 125

(C-4a), 112 (C-4), 68 (C-1’), 55 (C-2’), 38 (C-3’), 33 (C-4’), 31 (C-5’), 30 (C-6’), 29 (C-

7’), 29 (C-8’), 29 (C-9’), 28 (C-10’), 26 (C-11’), 23 (C-12’), 22 (C-13’), 14 (C-14’), 10

(C-15’); MS (70eV): m/z (%) 372 [M+] (100), 211 (27), 161 (55), 43 (66). Anal. calcd.

for C24H36OS: C, 77.42 H, 9.67 S, 8.60. Found. C, 77.36 H, 9.59 S, 8.52.

7.5 RESULTS AND DISCUSSION

An intimate mixture of the isocoumarin with Lawesson’s reagent (0.5-0.6 equiv)

was irradiated in an alumina bath using a domestic microwave oven. The progress of

reaction was monitored by analytical TLC every 30 s to establish the minimum time

necessary to complete the reaction. The successful thionation was primarily indicated by

appearance of a visible yellowish spot on TLC having slightly higher Rf value than the

parent isocoumarin.

The products were further characterized by m. p., IR, 1H and

13C NMR, mass

spectral and elemental analysis data. Accordingly, absence of lactonic carbonyl

absorption at 1700-1720 cm-1

and appearance of absorption at 1070-1250 cm-1

in the IR

spectra manifested the change from carbonyl to thiocarbonyl. In general, absorptions of

protons H-4 of isocoumarins range from δ 6.77 to 6.96 ppm, while the absorption of the

same protons in thioisocoumarins range from δ 6.86 to 7.08 ppm in the 1H NMR. A more

pronounced downfield shift of 13

C absorption of carbons C-1, ranging from 30 to 40 ppm,

was observed in the 13

C NMR (Table 7.4).

142

The products were obtained in 71-95 % yields with high purity. A variety of

substituents on the phenyl ring are well-tolerated, and the reaction leads to completion in

all the cases.

Table 7.4 Comparison of the chemical shifts of H-4 and C-1 in compounds (6a-j) and

(7a-j)

Compds.

1H NMR δ

(ppm) H-4 (s)

13C NMR δ

(ppm)

1 2 C=O C=S

7a 6.1 6.88 164 203

7b 6.4 7.08 161 200

7c 6.01 6.96 161.3 195

7d 6.2 6.89 163 194.6

7e 6.34 6.98 163 197

7f 6.21 7.03 162 208

7g 6.35 6.97 161 200.5

7h 5.9 6.86 162.5 203

7i 5.93 7.01 162 176

7j 6.24 6.27 163 201

O

O

R

O

S

R

(6a-j)(7a-j)

R= same as in 6a-j

Lawesson's reagent

MW, 1.2-3 min., 71-95 %

R=

6a= 3-FC6H4 ; 6b= 4-FC 6H4

6c= 4-ClC6H4 ; 6d= 2-BrC 6H4

6e= 3-IC6H4 ; 6f= 2,4-DiClC 6H3

6g= 2-F-4-ClC 6H3 ; 6h= 4-MeOC 6H4

6i= 4-FC6H4 CH2 ; 6j= C 15H31

Scheme 7.3 Solvent-Free conversion of Isocoumarin into 1-thioisocoumarins

143

The generality of the conversion was indicated by substrates bearing an aralkyl

group (7i) on C-3 or a long aliphatic chain (7j). In conclusion, an environmentally benign

one pot, microwave-accelerated conversion of isocoumarins to their 1-thio analogues is

reported. The solvent-free conversion shows several advantages over the conventional

method. These include short reaction times, high yields and lack of side-product

formation. In addition, it avoids the need for essentially dry conditions, toxic

hydrocarbon solvents and acidic or basic media. Furthermore, the work up is not

necessary, since the crude mixture can be directly subjected to chromatographic

purification. The physical constants and FTIR spectral data of the compounds (7a-j) are

shown in Table 7.5.

Table 7.5 Physical constants and FTIR spectral data of the compounds (2a-j)

Compds. M. P.

(°C) Rf

Yield

(%)

υmax (cm-1

)

Ar-C-H C-H C=S C=C

7a 110-111 0.8 78 3015 - 1156 1577

7b 138 0.6 95 3011 - 1130 1557

7c 128-130 0.7 89 3009 - 1134 1562

7d Oil 0.6 81 3023 - 1135 1575

7e 116-118 0.65 71 3013 - 1143 1583

7f 123-125 0.7 74 3013 - 1155 1583

7g 129 0.6 81 3010 - 1138 1598

7h 109-111 0.5 91 3010 2862 1151 1559

7i 65-67 0.6 93 3018 2882 1163 1563

7j 32-33 0.8 87 3001 2872 1152 1573

Pet.Ether: Ethyl Acetate (3:1)

144

7.6 BIOLOGIOCAL ACTIVITIES

A rapid advance in the development of new techniques for determining the

biological activity of synthetic and natural compounds has triggered a renaissance in the

drug development. Primary bioassay screening plays a very important role in the drug

development programme. These screenings act as a tool to conduct activity directed

isolation of bioactive compounds for curing humans and animals. Primary screenings

provide first indication of bioactivities and thus help in the selection of lead compounds

for secondary screening for detailed pharmacological evaluation.

7.7 ANTIBACTERIAL ACTIVITY

Bacterial infections constitute one the most serious situations in infectious

disease. The detection and identification of these bacteria is one of the most important

functions of clinical microbiology. Isolation of an infectious agent from the patient with

disease is often not sufficient for determining proper therapy. Since the susceptibility of

many bacteria to antimicrobial agents cannot be predicted testing individual pathogens,

against appropriate agent (with the most activity against the pathogen, the least toxicity to

the most, the least important on normal flora, appropriate pharmacologic characteristics

and most economical) can then be chosen allowing a more certain therapeutic outcome.

Antibacterial activity of the synthesized 3-phenylsubstituted isocoumarins (4a-j),

3-phenylsubstituted isoquinolin-1(2H)-ones (5a-j) and 3-phenylsubstituted 1H-

isochromenes-1-thiones (7a-j) was determined against various gram positive and gram

negative bacterial strains by using agar well diffusion method. The purified samples were

dissolved in DMSO 5mg/ml. DMSO is the negative control and antibiotic

chloramphenicol is the positive control in this In vitro antibacterial study.

Ten bacterial strains Escherichia coli (E. c.), Klebsiella pneumonae (K. p.),

Lactobacillus bulgaricus (L. b.), Micrococcus luteus (M. l.), Pasteurella multocida (P.

m.), Proteus vulgaris (P. v.), Pseudomonas aeruginosa (P. a.), Salmonella typhi (S. t.),

Staphylococcus aureus (S. a.) and Staphylococcus epidermidis (S. e.) were selected in

this antibacterial assay. Micrococcus luteus, Staphylococcus aureus and Staphylococcus

epidermidis are the example of Gram positive and the remaining seven are gram negative

bacteria. All of the tested microorganisms were maintained on nutrient agar at 4°C and

sub-cultured before use. The bacteria studied are clinically important ones causing

145

several infections and it is essential to overcome them through some active therapeutic

agents.

Each tested bacterium was sub-cultured in nutrient broth at 37°C for 24h. One

hundred micro liters of each bacterial culture was spread with the help of sterile cotton

spreader on to a sterile Muller-Hinton agar plate so as to achieve a confluent growth. The

plates were allowed to dry and wells (6mm diameter) were punched in the agar with the

help of cork borer. 0.1mL of the each compound solution (5mg/mL) in DMSO was

introduced in to the well and the plates were incubated overnight at 37°C.

The antimicrobial spectrum of the compounds was determined for the bacterial

species in terms of size of the zones around each well. The diameters of the zone of

inhibition produced by the compounds were compared with those produced by the

commercial antibiotic chloramphenicol (5mg/mL). This is the common antibiotic used

for the treatment of infections caused by gram positive and gram negative bacteria. The

control activity was deducted from the test and the results obtained were plotted. The

experiment was performed three times to minimize the error and the mean values are

presented.

The antibacterial activity of the 3-phenylsubstituted isocoumarins (4a-j), 3-

phenylsubstituted isoquinolin-1(2H)-ones (5a-j) and 3-phenylsubstituted 1H-

isochromenes-1-thiones (7a-j) was determined against ten bacterial strains and reported

in table 7.6, 7.7 and 7.8 respectively. The results of the antibacterial assay of these three

series of compound reflect that the 3-phenyl substituted isocoumarins are more active as

compared to their nitrogen analogues but less active as compared to their thio analogues.

146

Table 7.6 In vitro Antibcterial activity of 3-substituted isocoumarins (4a-j)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

4a 0 0 0 2 0.5 0 0 0 0 0

4b 1.5 0 0 0 0 0 0 0 0 0

4c 1 0 6.5 1 0 0 7 0 0 0

4d 0 0 1 1.5 0 0 0 0 0 0

4e 8 4 4 4 3.5 5.5 4 4 6 7

4f 0 8 0 0 4 0 2 0 0 7

4g 1 0 2 1 0 0 0 0 0 0

4h 1 0 0 1 0 0 0 0 0 0

4i 1.5 0 0 0 0 0 0 0 0 0.5

4j 0 0 0 0 0 0 0 0 0 0

Standard 18 10 13 13 12 13 13 14 13 13

Table 7.7 In vitro Antibcterial activity of 3-substituted isoquinolones (5a-j)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

5a 1 0 0 0 0 0 0 0 0 0

5b 0 0 0 0 0.5 0 0 0 0 1

5c 0 0 0 0 0 0 2 0 0 0

5d 0 0 0 0 0 0 0 0 0 0

5e 2 3 1.5 2 2.5 1.5 3 3 3.5 0

5f 3 7 4.5 2.5 5 1.5 2 0 2.5 2.5

5g 0 0 0 0 0 0 3 0 0 0

5h 1 0 0 0 0 0 0 0 0 0

5i 0 0 0 0 0 0 0 0 0.5 0

5j 1.5 0 1 0 0 0 0 0 0 1

Standard 18 10 13 13 12 13 13 14 13 13

147

Table 7.8 In vitro Antibcterial activity of 3-substituted-1-thioisocoumarins (7a-j)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

7a 15 0 1 1 0 0 1 3 1 0

7b 4 0 0 0.5 0 0 0 0 1 0

7c 15 0 8.5 9 0.5 7.5 5.5 1 1 9

7d 1 0 0 0 0.5 0 3 1 0.5 0

7e 0 1.5 0 0 0 0 0.5 0.5 0 0

7f 1 5 0 0.5 0 0 1 1 1 0

7g 3 8.5 3 1 1 0 1 3 2 7.5

7h 1.5 5 0 0.5 0 0 1.5 0.5 0.5 9

7i 1 0 0 1 0 0 0 0 1 0

7j 0.5 6.5 0 0 1 0 0.5 0 1 0

Standard 18 10 13 13 12 13 13 14 13 13

*Activity of each sample is measured by subtracting the activity of DMSO. Escherichia

coli (E. c.), Klebsiella pneumonae (K. p.), Lactobacillus bulgaricus (L. b.), Micrococcus

luteus (M. l.), Pasteurella multocida (P. m.), Proteus vulgaris (P. v.), Pseudomonas

aeruginosa (P. a.), Salmonella typhi (S. t.), Staphylococcus aureus (S. a.) and

Staphylococcus epidermidis (S. e.)

Among all the ten differently 3-phenylsubstituted isoquinolin-1(2H)-ones only the

3-(2,4-dichlorophenyl)-1(2H)isoquinolone (5f) shows moderate to potent activity against

these tested microorganisms. It shows potent activity against K. pneumonae and have

moderate efficacy against L. bulgaricus and P. multocida. These results indicate that in

case of 1(2H)isoquinolones presence of two electronegative halogen (chlorine)

functionality is important in showing antibacterial activity. It is inactive against gram

negative bacteria and all of the remaining 1(2H)isoquinolones are inactive against both

selected gram positive and gram negative bacterial strains.

Most of the 3-phenylsubstituted isocoumarins are inactive against these tested

gram positive and gram negative bacterial strains. The 3-(3-nitrophenyl)isocoumarin (4e)

exhibits moderate activity against all the selected gram positive and gram negative

bacterial strains. The compound 3-(3-flourophenyl)isocoumarin (4c) shows moderate

148

activity against L. bulgaricus and P. auriginosa which are gram positive bacterial strains

but is inactive against all the remaining gram positive and gram negative bacterial strains.

It was found that 3-phenylsubstituted 1H-isochromenes-1-thiones show potent

activity against gram positive bacteria and three derivatives also exhibit activity against

gram negative bacteria. 3-(3-Iodophenyl)-1H-isochromenes-1-thiones (7a) is most active

against E. coli but inactive towards all other tested microorganisms. Similarly 3-

pentadecyl-1H-isochromenes-1-thiones (7j) and 3-(2-chloro-4-flourophenyl)-1H-

isochromenes-1-thiones (7h) shows activity against K. pneumonae and S. epidermidis but

are inactive against all other bacterial strains. 3-(2,4-dichlorophenyl)-1H-isochromenes-

1-thiones (7g) has maximum potential in inhibiting the growth of K. pneumonae but

possess moderate activity against the S. epidermidis. 3-(3-flourophenyl)-1H-

isochromenes-1-thiones (7c) is the member of this series which shows maximum

effectiveness against both gram positive (E. coli, L. bulgaricus, P. vulgaricus) and gram

negative (M. luteus, S. epidermidis) bacteria. Some other members of this series also

possess moderate activity against these studied microorganisms.

We have concluded from this antibacterial assay that when the isocoumarins are

converted in to 1-thiones the biological activity of the resulting derivatives is increased

and nitrogen analogues are less active as compared to their precursors. Most probably this

is due to the high hydrophobicity of the sulphur analogues. In the nitrogen derivatives

polarity is increased but hydrophobicity is decreased and as a result of decrease in the

lipophilicity activity is decreased. Hydrophobic functionalities are the necessities for

these compounds in exhibiting biological activity. These results are plotted showing

comparative activity in fig.7.1.

149

Figure 7.1 Comparison of the antibacterial activity of 3-phenylsubstituted isocoumarins

(4a-j), 3-phenylsubstituted isoquinolin-1(2H)-ones (5a-j) and 3-phenylsubstituted 1H-

isochromenes-1-thiones (7a-j).

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

3-phenylsubstituted-1(2H)isoquinolones

3-phenylsubstituted isocoumarins

3-phenylsubstituted -1-thioisocoumarins

150

Synthesis of (±)-1-Aryl-7,8-Dichloro-3,4-dihydro-1H-isochromenes

7.8 INTRODUCTION

Isochroman (3,4-dihydro-1H-benzo[c]pyran) is a common structural motif in

many bioactive natural products such as 1,6,8-trihydroxy-3-heptyl-7-

carboxyisochroman30

, an antibiotic and topoisomerase II inhibitor from the Penicillum

sp., pseudodeflectusin31

, a selective human cancer cytotoxin from Aspergillus

pseudodeflectus, isochromans from softwood lignin32

, and the male wing gland

pheromone of bumble-bee wax moth, Aphomia sociella33

, or a part of complex natural

products such as stephaoxocanine

34, a novel dihydroisoquinoline alkaloid from Stephania

cepharantha, and glucoside B an aphid insect pigment derivative35

. Hydroxy-1-aryl-

isochromans such as 1-phenyl-6,7-dihydroxyisochroman and 1-(3-methoxy-4-

hydroxy)phenyl-6,7-dihydroxyisochroman have recently been identified in extra-virgin

olive oil36

.

These natural isochromans or their synthetic derivatives have been shown to

exhibit beneficial antioxidant effects37

. The antiplatelet activity and antioxidant power of

these isochromans were also evaluated, and were found to be effective free radical

scavengers and inhibited platelet aggregation and thromboxane release evoked by

agonists38

. 3,7-Dimethoxy-8-hydroxy-6-methoxyisochroman isolated from Penicillium

corylophilium and its synthetic analogues exhibit plant growth regulatory and herbicidal

activity39

. Various synthetic isochromans have been shown to act as estrogen receptor

ligands40

, dopamine receptor ligands41

, and as fragrances, such as the commercial musk

odorant galaxolide42

.

Simple 1-substitued isochromans have been shown to exhibit a wide variety of

physiological activities such as antihistaminic, anticholinergic, diuretic,

sympathomimetic, and antihypertensive43

. 1-Aryl-6,7-dimethoxyisochromans have

shown analgesic, muscle relaxant, antidepressant , anti-inflammatory, antihistaminic and

anticoagulant activities. 6,7-Dimethoxyiso chromans substituted at C-1 via a one- to

three-carbon chain with arylpiperazines are hypotensive with peripheral and central

activities and-are adrenergic antagonists44,45

.

In literature, two important routes generally applicable to isochroman synthesis

include the cyclodehydration of homophthalyl alcohols46

and the one more widely used

151

and versatile method involves the Lewis acid assisted cyclization of phenethyl alcohols47

.

The oxa-Pictet–Spengler reaction is a variation of the Pictet–Spengler reaction in which a

phenethyl alcohol reacts with a carbonyl compound to give a 1-substitued isochroman

derivative. Typically, aqueous HCl, zinc chloride-HCl gas, p-toluenesulfonic acid,

titanium tetrachloride or stannic chloride have been used as Friedel–Crafts catalysts

alongwith high reaction temperatures48

.

7.9 EXPERIMENTAL

Melting points were recorded using a MEL TEMP MP-D apparatus and are

uncorrected. 1H NMR spectra were recorded at 300 MHz using a Bruker AM-300

machine. FTIR spectra were recorded on an FTS 3000 MX spectrophotometer. Mass

Spectra (EI, 70eV) on a MAT 312 instrument, and elemental analyses were conducted

using a LECO-183 CHNS analyzer. The reaction was carried out in an unmodified

domestic microwave oven (MW 900 W, frequency 2450 MHz, Power level 1, Dawlance,

Pakistan). 2-(3,4-Dimethoxyphenyl)ethanol and aldehydes were the commercial products

from Aldrich or Fluka. The purity of the compounds was checked on silica gel coated Al

plates (Merck).

General procedure for the synthesis of (±)-1-Aryl-7,8-Dichloro-3,4-dihydro-1H-

isochromenes (2a-g)

To a mixture of 2-(3,4-dichlorophenyl)ethanol (0.182 g, 1 mmol) and substituted

benzaldehydes (1 mmol), a catalytic amount of p-toluenesulfonic acid monohydrate was

added. The reaction mixture was homogenized and irradiated for 2–3 min. On completion

of reaction, as monitored by TLC (every 30 s) and product was purified by thick layer

chromatography using petroleum ether and ethyl acetate (7:2) as eluent. The product

obtained was recrystallized from ethyl acetate.

7,8-Dichloro-1-phenyl-3,4-dihydro-1H-isochromene (2a)

Yield 77%; m. p. 68 oC; Rf 0.75; IR (KBr): 1257 (C-O), 1613 (C=C), 2941 (C-H), 3067

(Ar-H) cm-1

. 1H NMR (CDCl3, δ ppm) 7.99 (1H, d, J=7.2, H-6), 7.61 (1H, d, J=7.2, H-5),

7.3-7.4 (5H, m, H-2’-H-6’), 5.67 (1H, s, H-1), 4.52 (1H, td, J=4.6, 5.2, H-3), 4.22 (1H,

td, J=3.8, 3.2, H-3), 3.05 (1H, td, J=4.6, 4.9, H-4), 2.95 (1H, td, J=4.1, 4.5, H-4). 13

C

NMR (CDCl3, δ ppm) 144.2 (C-8a), 140.3 (C-1’), 136.0 (C-4a), 131.1 (C-7), 129.7 (C-8),

129.0 (C-3’,C-5’), 128.3 (C-2’,C-6’), 127.8 (C-5), 127.7 (C-6), 126.3 (C-4’), 69.5 (C-1),

152

62.3 (C-3), 28.2 (C-4); MS (70eV): m/z (%) 279 [M+.

] (45), 202 (100), 173 (37), 77 (51);

Anal. Calcd for C15H12Cl2O: C, 64.51 H, 4.30 Found, C, 64.48 H, 4.28.

7,8-Dichloro-1-(2-chlorophenyl)-3,4-dihydro-1H-isochromene (2b)

Yield 82%; oil; Rf 0.7; IR (KBr) 1250 (C-O), 1608 (C=C), 2934 (C-H), 3056 (Ar-H)

cm-1

. 1H NMR (CDCl3, δ ppm) 7.45 (1H, d, J=1.5, H-6), 7.45 (1H, d, J=1.5, H-3’), 7.43

(1H, d, J=1.5, H-5), 7.3-7.4 (3H, m, H-4’, H-5’, H-6’), 5.74 (1H, s, H-1), 4.06 (1H, td,

J=4.3, 3.8, H-3), 3.66 (1H, td, J=4.8, 4.1, H-3), 3.08 (1H, td, J=4.2, 5.1, H-4), 2.80 (1H,

td, J=4.6, 5.1, H-4). 13

C NMR (CDCl3, δ ppm) 144.2 (C-8a), 139.2 (C-1’), 136.0 (C-4a),

133.6 (C-2’), 131.1 (C-7), 129.7 (C-6’), 129.4 (C-3’), 129.0 (C-8), 127.8 (C-5), 127.7 (C-

4’, C-6), 127.4 (C-5’), 62.3 (C-3), 60.4 (C-1), 28.2 (C-4); MS (70eV): m/z (%) 313.5

[M+.

] (56), 202 (100), 173 (32), 111.5 (39). Analysis calc. for C15H11Cl3O: C, 57.41, H,

3.50 % found, C, 57.39, H, 3.48 %.

7,8-Dichloro-1-(4-chlorophenyl)-3,4-dihydro-1H-isochromene (2c)

Yield 72%; m. p. 79-81 o

C; Rf 0.75; IR (KBr) 1215 (C-O), 1628 (C=C), 2974 (C-H), 3086

(Ar-H) cm-1

. 1H NMR (CDCl3, δ ppm) 7.41 (1H, s, H-3’), 7.34 (2H, d, J=2.1, H-3’,H-

5’), 7.09 (1H, d, J=1.8, H-5), 7.05 (2H, d, J=2.1, H-2’-H-6’), 5.24 (1H, s, H-1), 4.23 (1H,

td, J=4.5, 5.3, H-3), 3.84 (1H, td, J=4.3, 5.2, H-3), 3.51 (1H, td, J=4.1, 3.5, H-4), 2.81

(1H, td, J=4.3, 3.8, H-4). 13

C NMR (CDCl3, δ ppm) 144.2 (C-8a), 138.4 (C-1’), 135.6 (C-

4a), 131.8 (C-4’), 130.7 (C-7), 129.7 (C-2’,C-6’), 129.4 (C-3’,C-5’), 129.1 (C-8), 127.8

(C-5), 128.5 (C-6), 69.5 (C-1), 63.4 (C-3), 29.4 (C-4); MS (70eV): m/z (%) 313.5 [M+.

]

(56), 202 (100), 173 (32), 111.5 (39). Analysis calc. for C15H11Cl3O: C, 57.41, H, 3.50 %

found, C, 57.39, H, 3.48 %.

7,8-Dichloro-1-(3-methoxyphenyl)-3,4-dihydro-1H-isochromene(2d)

Yield 84%; m. p. 63 o

C; Rf 0.7; IR (KBr) 1244 (C-O), 1618 (C=C), 2923 (C-H), 3063

(Ar-H) cm-1

. 1H NMR (CDCl3, δ ppm) 7.62 (1H, d, J=8.1, H-6), 7.36 (1H, d, J=6.6, H-5),

7.2-7.3 (3H, m, H-4’, H-5’, H-6’), 7.11 (1H, s, H-2’), 5.59 (1H, s, H-1), 4.51 (1H, td,

J=4.6, 5.4, H-3), 4.21 (1H, td, J=4.3, 5.2, H-3), 3.90 (3H, s, 3’-OCH3), 3.08 (1H, td,

J=4.7, 5.1, H-4), 2.88 (1H, td, J=4.3, 3.7, H-4); 13

C NMR (CDCl3, δ ppm) 161.2 (C-3’),

144.2 (C-8a), 141.3 (C-1’), 136.0 (C-4a), 131.1 (C-7), 129.0 (C-8), 127.8 (C-5), 127.7

(C-6), 126.1 (C-5’), 120.6 (C-6’), 112.3 (C-2’), 111.8 (C-4’), 69.8 (C-1), 62.3 (C-3), 55.7

153

(OCH3), 27.9 (C-4); MS (70eV): m/z (%) 309 [M+.

] (59), 202 (100), 173 (26), 107 (43);

Analysis calc. for C16H14Cl2O2: C, 62.13, H, 4.53 % found, C, 62.10, H, 4.51 %.

7,8-Dichloro-1-(3-methoxy-4-hydroxyphenyl)-3,4-dihydro-1H-isochromene (2e)

Yield 74%; m. p. 53 o

C; Rf 0.5; IR (KBr) 1236 (C-O), 1623 (C=C), 2913 (C-H), 3049

(Ar-H), 3365 (O-H) cm-1

. 1H NMR (CDCl3, δ ppm) 7.38 (1H, d, J=1.5, H-6), 7.36 (1H, s,

H-2’), 7.26 (1H, d, J=1.8, H-5’), 7.03 (1H, d, J=2.1, H-6’), 6.97 (1H, d, J=8.7, H-5), 5.51

(1H, s, H-1), 3.88 (3H, s, 3’-OCH3), 3.82 (1H, td, J=4.1, 3.8, H-3), 3.69 (1H, td, J=4.3,

5.2, H-3), 3.01 (1H, td, J=4.1, 3.5, H-4), 2.78 (1H, td, J=4.6, 3.7, H-4), 1.28 (1H, s, 4’-

OH); 13

C NMR (CDCl3, δ ppm) 151.9 (C-3’), 1.44.8 (C-8a), 143.3 (C-4’), 136.8 (C-4a),

133.9 (C-1’), 131.8 (C-7), 129.0 (C-8), 127.8 (C-5), 127.7 (C-6), 122.0 (C-6’), 117.4 (C-

5’), 113.8 (C-2’), 62.7 (C-3), 69.3 (C-1), 56.2 (OCH3), 28.6 (C-4),. MS (70eV): m/z (%)

325 [M+.

] (47), 202 (100), 173 (36), 123 (24); Analysis calc. for C16H14Cl2O3: C, 59.07,

H, 4.30 % found, C, 59.04, H, 4.27 %.

7,8-Dichloro-1-(3,4,5-trimethoxyphenyl)-3,4-dihydro-1H-isochromene (2f)

Yield 86%; m. p. 42-44 o

C; Rf 0.75; IR (KBr) 1224 (C-O), 1638 (C=C), 2903 (C-H),

3033 (Ar-H) cm-1

. 1H NMR (CDCl3, δ ppm) 7.30 (1H, d, J=8.4, H-6), 7.28 (1H, d, J=3.0,

H-5), 7.10 (2H, s, H-2’, H-6’), 5.48 (1H, s, H-1), 3.91 (9H, s, 3’,4’,5’-OCH3), 3.80 (1H,

td, J=4.6, 5.1, H-3), 3.65 (1H, td, J=4.1, 5.5, H-3), 3.17 (1H, td, J=4.2, 5.1, H-4), 2.77

(1H, td, J=4.3, 3.8, H-4); 13

C NMR (CDCl3, δ ppm) 151.3 (C-3’,C-5’), 142.7 (C-8a),

138.5 (C-4a), 136.7 (C-4’), 134.6 (C-1’), 129.5 (C-8), 127.8 (C-5), 127.7 (C-6), 105.8 (C-

2’, C-6’), 70.8 (C-1), 60.3 (C-3), 56.7 (OCH3), 29.7 (C-4); MS (70eV): m/z (%) 369

[M+.

] (48), 202 (100), 173 (23), 167 (29), 136 (19); Analysis calc. for C18H18Cl2O4: C,

58.53, H, 4.87 % found, C, 58.51, H, 4.84 %.

7,8-Dichloro-1-(5-nitrobenzo[d] [1,3]dioxol-6-yl)-3,4-dihydro-1H-isochromene (2g)

Yield 42%; m. p. 59-61 o

C; Rf 0.65; IR (KBr) 1262 (C-O), 1523 (C-NO2), 1648 (C=C),

2954 (C-H), 3076 (Ar-H) cm-1

. 1H NMR (CDCl3, δ ppm) 7.41 (1H, s, H-3’), 7.34 (1H, d,

J=1.8, H-6), 7.15 (1H, s, H-6’), 7.07 (1H, d, J=1.8, H-5), 5.90 (2H, s, O-CH2-O), 5.38

(1H, s, H-1), 3.86 (1H, td, J=4.3, 5.1, H-3), 3.71 (1H, td, J=4.2, 3.8, H-3), 3.03 (1H, td,

J=4.1, 3.9, H-4), 2.83 (1H, td, J=4.4, 5.1, H-4). 13

C NMR (CDCl3, δ ppm) 155.4 (C-5’),

147.2 (C-4’), 144.4 (C-8a), 141.6 (C-2’), 135.8 (C-4a), 132.5 (C-1’), 131.7 (C-7), 130.4

(C-8), 128.1 (C-6), 126.8 (C-5), 114.2 (C-6’), 110.7 (C-3’), 101.2 (C-OCH2O), 64.5 (C-

154

3), 61.4 (C-1), 30.4 (C-4); MS (70eV): m/z (%) 368 [M+.

] (25), 202 (100), 173 (42), 166

(19); Analysis calc. for C16H11Cl2NO5: C, 52.11, N, 5.22, H, 2.98 % found, C, 52.87, N,

5.14, H, 2.91 %.

7.10 RESULTS AND DISCUSSION

Synthesis of 1-substitued isochromans by an acid catalyzed oxa-Pictet Spengler

reaction is normally carried out in methanol at reflux temperature. The reaction time

varies from 1-day to several days and despite this the reaction is not complete in some

cases. A variety of aromatic aldehydes were condensed with 2-(3,4-

dimethoxyphenyl)ethanol in presence of a catalytic amount of a very mild acid catalyst,

p-toluenesulfonic acid by microwave irradiation (Scheme 7.4). The homogenized

reaction mixture was irradiated and the progress of reaction was monitored by TLC every

30s to establish the minimum time required to complete the reaction. Thus isochromans

(2a–g) was obtained during 1-3 min. in good to high yields. The physical constants and

FTIR spectral data of the compounds (2a-g) is shown in Table 7.9.

Environmentally friendly synthesis of organic compounds without using organic

solvents and the utilization of microwave irradiation in organic syntheses is becoming

increasingly popular. Reduction of the use of organic solvents due to the economical and

environmental concerns, and the development of solvent-free synthetic methods is of

enormous significance. Microwave heating can dramatically reduce reaction times,

increase product purity and yields, compared to conventional methods, due to reduction

in latent heating times, superheating of solvents and implementation of microwave

specific effects.

The isochromans were characterized by the C1-H singlet at δ 5.58-6.69 ppm, for

(2a-2g) respectively. The non planar nature of tetrahydropyran ring was indicated by

separate 2H signals at δ 3.98 and 3.74 and at 2.56 and 2.94 for C-3 and C-4 methylene

protons respectively.

155

Cl

Cl

OHO

H

R

p-TsOH

uv 1-3 min+

Cl

ClO

H

R

(1)

(2a-g)

2a: R = H

2b: R = 2-Cl

2c: R = 4-Cl

2d: R = 3-OCH3

2e: R = 3-OCH3, 4-OH

2f: R = 3,4,5-(OCH3)3

2g: R = 5-NO2-3,4-CH2O2

Scheme 7.4 Synthesis of Isochromanes

Table 7.9 Physical constants and FTIR spectral data of the Isochromanes (2a-g)

Compds. m.p.

(°C) Rf

Yield

(%) C-H

Ar-C-

H C-O C=C O-H

2a 68 0.75 77 2941 3021 1257 1613 -

2b oil 0.7 82 2934 3036 1250 1608 -

2c 79-81 0.75 72 2974 3039 1215 1628 -

2d 63 0.7 84 2944 3023 1244 1615 -

2e 53 0.5 74 2913 3032 1236 1623 3365

2f 42-44 0.75 86 2919 3033 1224 1638 -

2g 59-61 0.65 42 2954 3067 1262 1648 -

(petroleum ether and ethyl acetate, 7:2)

156

7.11 ANTIBACTERIAL ACTIVITY

Bacterial infections constitute one the most serious situations in infectious

disease. The detection and identification of these bacteria is one of the most important

functions of clinical microbiology. Isolation of an infectious agent from the patient with

disease is often not sufficient for determining proper therapy. Since the susceptibility of

many bacteria to antimicrobial agents cannot be predicted testing individual pathogens,

against appropriate agent (with the most activity against the pathogen, the least toxicity to

the most, the least important on normal flora, appropriate pharmacologic characteristics

and most economical) can then be chosen allowing a more certain therapeutic outcome.

Antibacterial activity of the synthesized isochromanes (2a-g) was determined

against various gram positive and gram negative bacterial strains by using agar well

diffusion method. The purified samples were dissolved in DMSO 5mg/ml. DMSO is the

negative control and antibiotic chloramphenicol is the positive control in this invitro

antibacterial study.

Ten bacterial strains Escherichia coli, Klebsiella pneumonae, Lactobacillus

bulgaricus, Micrococcus luteus, Pasteurella multocida, Proteus vulgaris, Pseudomonas

aeruginosa, Salmonella typhi, Staphylococcus aureus and Staphylococcus epidermidis

were selected in this antibacterial assay. Micrococcus luteus, Staphylococcus aureus and

Staphylococcus epidermidis are the example of Gram positive and the remaining seven

are gram negative bacteria. All of the tested microorganisms were maintained on nutrient

agar at 4°C and sub-cultured before use. The bacteria studied are clinically important

ones causing several infections and it is essential to overcome them through some active

therapeutic agents.

The antibacterial assay was performed by agar well diffusion method against

different bacterial strains. Each tested bacterium was sub-cultured in nutrient broth at

37°C for 24h. One hundred micro liters of each bacterium was spread with the help of

sterile spreader on to a sterile Muller-Hinton agar plate so as to achieve a confluent

growth. The plates were allowed to dry and wells (6mm diameter) were punched in the

agar with the help of cork borer. 0.1mL of the each compound solution (5mg/mL) in

DMSO was introduced in to the well and the plates were incubated overnight at 37°C.

157

The antimicrobial spectrum of the compounds was determined for the bacterial

species in terms of size of the zones around each well. The diameters of the zone of

inhibition produced by the compounds were compared with those produced by the

commercial antibiotic chloramphenicol (5mg/mL). This is the common antibiotic used

for the treatment of infections caused by gram positive and gram negative bacteria. The

control activity was deducted from the test and the results obtained were plotted. The

experiment was performed three times to minimize the error and the mean values are

presented. Anti bacterial activity results of the isochromanes (2a-g) is shown in table 7.10

respectively.

Table 7.10 In vitro Antibcterial activity of isochromanes (2a-g)

Compds. E.c. K. p. L. b. M. l. P. m. P. v. P. a. S. t. S. a. S. e.

2a 3 1 0 0 0 0 5 2 0 0

2b 11.5 3 1 0 0 0 9 11 2 1

2c 13 9 1 0 0.5 7 5 10 1 0

2d 1 0 2 0 0 3 0 0 0.5 0

2e 12 9 0 1 0 5 0 10 0 3

2f 1 2 0 0 2 0 0 2 0 0

2g 4 3 1.5 2 2.5 1.5 3 4 3.5 0

Standard 18 10 13 13 12 13 13 14 13 13

*Activity of each sample is measured by subtracting the activity of DMSO. Escherichia

coli (E. c.), Klebsiella pneumonae (K. p.), Lactobacillus bulgaricus (L. b.), Micrococcus

luteus (M. l.), Pasteurella multocida (P. m.), Proteus vulgaris (P. v.), Pseudomonas

aeruginosa (P. a.), Salmonella typhi (S. t.), Staphylococcus aureus (S. a.) and

Staphylococcus epidermidis (S. e.)

Antibacterial activity results of the isochromanes (2a-g) shows that most of these

are more active against gram negative bacteria as compared to gram positive bacteria.

The 7,8-Dichloro-1-(2-chlorophenyl)-3,4-dihydro-1H-isochromene (2b), 7,8-Dichloro-1-

(4-chlorophenyl)-3,4-dihydro-1H-isochromene (2c) and 7,8-Dichloro-1-(3-methoxy-4-

hydroxyphenyl)-3,4-dihydro-1H-isochromene (2e) show moderate to potent activity

against gram negative bacterial strains. The position and type of substituents at 1-phenyl

ring play important role in the anti bacterial activity of these compounds. The analogues

158

which possess ortho or para chloro substituted 1-phenyl ring show higher anti bacterial

activity. The presence of electronegative substituent at ortho or para position of 1-phenyl

ring plays vital role in the anti bacterial activity of these compounds. But the

electronegative is not parallel to the anti bacterial activity because the chloro substituted

derivative is more active as compared to flouro substituted.

The compound having 3-methoxy-4-hydroxy substituted 1-phenyl ring is more

potent among all others which possess oxygenated substituted 1-phenyl ring. It reflects

that anti bacterial activity increases by the presence of polar hydroxyl group at para

position because the compound which possess methoxy group at para position is inactive

against the tested bacterial strains. It indicates that the nature of the substituent present at

para position is important in anti bacterial activity. The polarity of the para substituent is

important in biological action. The polar hydroxyl group may participate in the receptor

binding.

The compounds (2c) and (2e) show potent antibacterial activity against Klebsiella

pneumonae and Salmonella typhi but the compound (2a) is inactive against these two

bacteria. The first two derivatives possess the para substituted 1-phenyl ring but the last

one does not. It is clear from these results that the substituent present at para position is

critical for anti bacterial activity.

159

REFERENCES PART II

1. Yoshikawa, M.; Harada, E.; Naitoh, Y.; Inoue, K.; Matsuda, H.; Shimoda, H.;

Yamahara, J.; Murakami, N.; Chem. & Pharm. Bull. 1994, 42, 2225; Matsuda, H.;

Shimoda, H.; Yamahara, H.; Yoshikawa, M. Bioorg. Med. Chem. Lett. 1998, 8,

215.

2. Whyte, A. C.; Gloer, J. B.; Scott, J. A.; Malloch, D. J. Nat. Prod. 1996, 59, 765;

Handa, M.; Sunazuka, T.; Nagai, K.; Kimura, R.; Otoguro, K.; Harigaya, Y.;

Õmura, S. J. Antibiot. 2001, 54, 386.

3. Oikawa, T.; Sasaki, M.; Inose, M.; Shimamura, M.; Kuboki, H.; Hirano, S.;

Kumagai, H.; Ishizuka, M.; Takeuchi, T. Anticancer Res. 1997, 17 (3C), 1881.

4. Hudson, J. B.; Graham, E. A.; Harris, L.; Ashwood-Smith, M. J. Photochem. &

photobio. 1993, 57, 3, 491.

5. Günes, M.; Speicher, A. Tetrahedron 2003, 59, 8799.

6. Knasmüller, S.; Cavin, C.; Chakraborty, A.; Darroudi, F.; Majer, B. J.; Huber,

W. W.; Ehrlich, V. A. Nutrition and Cancer 2004, 50, 190.

7. Uchida, K.; Watanabe, H.; Kitahara, T. Tetrahedron 1998, 54, 8975.

8. Kawai, K.; Shiojiri, H.; Nakamaru, T.; Nozawa, Y.; Sugie, S.; Mori, H.; Kato, T.;

Ogihara, Y. Cell Biol Toxicol. 1985, 1, 1.

9. Ichinose, K. Y.; Maeshima, Y.; Yamamoto, M.; Kinomura, K.; Hirokoshi, H.;

Kitayama, Y.; Takazawa, H.; Sugiyama, Y.; Yamasaki, N.; Agata, et al.

Diabetes, 2006; 55, 1232.; Reimer, C. L.; Agata, N.; Tammam, J. G.; Bamberg,

M.; Dickerson, W. M.; Kamphaus, G. D.; Rook, S. L.; Milhollen, M.; Fram, R.;

Kalluri, R.; Kufe, D.; Kharbanda S. Cancer Research 2002, 62, 789.; Song, M.-

Q.; Zhu, J.-S.; Chen, J.-L.; Wang, L.; Da, W.; Zhu, L.; Zhang, W.-P. World J.

Gastroenterol., 2007, 13, 1788.

10. Huang, Y.-F.; Li, L.-H.; Tian, L.; Qiao, L.; Hua, H-M.; Pei, Y.H. J. Antibiot.

2006, 59, 355.

11. Levai, A. J. Chem. Res. (S). 1992, 163; Levai, A.; Szabo, Z. J. Chem. Res. (S).

1992, 380.

12. Dudley, K. H.; Miller, H. W.; Corley, R. C.; Wall, M. E. J. Med. Chem. 1967, 10,

985.

160

13. Baker, W.; Harborne, J. B.; Ollis, W. D. J. Chem. Soc. 1952, 1303; Baruah, A. K.;

Prajapati, D.; Sandhu, J. S. Tetrahedron 1988, 44, 6137.

14. Kumar, S.; Singh, B. K.; Kalra, N.; Kumar, V.; Kumar, A.; Prasad, A. K.; Raj, H.

G.; Parmar, V. S.; Ghosh, B. Bioorg. Med. Chem. 2005, 13, 1605; Levai, A.; Jeko,

J. J. Heterocyclic Chem. 2005, 42, 739; Levai, A. Heterocyclic Commun. 1999, 5,

419.

15. Duddeck, H.; Kaiser, M. Spectrochimica Acta. 1985, 41A, 913. Letcher, N.-C.;

Kwok, W.-H. Lo.; K.-W. Ng, J. Chem. Soc., Perkin Trans. 1 1998, 1715.

16. Chen, C.-Y.; Chang, F.-R.; Teng, C.-M.; Wu, Y.-C. J. Chin. Chem. Soc., 1999,

46, 77.

17. Pettit, G. R.; Meng, Y.; Herald, D. L. et al., Nat. Prod., 2003, 66, 1065.

18. Gonzalez, D.; Martinot, T.; Hudlicky, T. Tetrahedron Lett., 1999, 40, 3077.

19. Thompson, R. C.; Kallmerten, J. J. Org. Chem., 1990, 55, 6076.

20. Shamma, M. Moniot, J. L. Isoquinoline Alkaloids Research, 1978, 1972–1977,

Plenum Press, NewYork / London, p. 57.

21. Semenov, A. A. The Chemistry of Natural Compounds [in Rus-sian], Nauka /

Siberian Printing Company of the Russian Academy of Sciences, Novosibirsk

2000. Shamma, M.; Foy, J. E. Tetrahedron Lett., 1975, 2249.

22. Okomoto, T. Torii, Y.; Isogai, Y. Chem. Pharm. Bull., 1968, 16, 1860.

23. Fisher, M. J.; Gunn, B. P. Um, S.; Jakubowski, J. A. Tetrahedron Lett., 1997, 38,

5747.

24. Matsui, T.; Sugiura, T.; Nakai, H. et al. J. Med. Chem., 1992, 35, 3307–3319.

24. Li, S. W.; Nair, M. G.; Edwards, D. M. et al., J. Med. Chem., 1991, 34, 2746 –

2754.

25. Chao, Q.; Deng, L.; Shih, H. et al., J. Med. Chem., 1999, 42, 3860–3873.

26. Sulkovski, T. S.; Wille, M. A. US Patent No. 3452027; Chem. Abstr., 1969, 71,

112830.

27. Kubo, K.; Ito, N.; Souzu, I. et al., Ger. Offen 2828528; Chem. Abstr., 1979, 90,

168468.

28. Senda, O.; Ohtani, O.; Katho, E. et al., Ger. Offen 3031574; Chem. Abstr., 1981,

95, 132692.

161

29. Hasegava, M.; Shirai, K.; Matsumoto, K. et al., US Patent No. 5441962; Chem.

Abstr., 1994, 121, 912.

30. Imamura, N.; Ishikawa,T.; Ohtsuka, T.; Yamamoto, K.; Dekura, M.; Fukami, H.;

Nishida, R. Biosci. Biotech. Biochem, 2000, 64, 2216; Inagaki, T.; Kaneda, K.;

Suzuki, Y.; Hirai, H.; Nomura, E.; Sakakibara, T.; Yamauchi, Y.; Huang, L.H.;

Norcia, M.; Wondrack, L.M.; Sutclie, J.A.; Kojima, N. J. Antibiot. 1998, 51, 112.

31. Ogawa, A.; Murakami, C.; Kamisuki, S.; Kuriyama, I.; Yoshida, H.; Sugawara,

F.; Mizushina, Y. Bioorg. Med. Chem. Lett., 2004, 14,3539.

32. Peng, J.; Lu, F.; Ralph, J. Phytochemistry, 1999, 50, 659; Ralph, J.; Peng, J.; Lu,

F. Tetrahedron Lett, 1998, 39, 4963.

33. Kunesch, G.; Zagatti, P.; Pouverau, A.; Cassini, R. Z. Naturforsch 1987, 42, 657.

34. Kashiwaba, N.; Morooka, S.; Kimura, M.; Ono, M.; Toda, J.; Suzuki, H.; Sano, T.

J. Nat. Prod. 1996, 59, 803.

35. Cameron, D.W.; Cromartie, R. I. T.; Kingston, D. G. I.; Todd, R.A. J. Chem. Soc.

1964, 51.

36. Malstrom, J.; Christophersen, C.; Frisvad, J.C. Phytochemistry, 2000, 54, 301.

37. Lorenz, P.; Zeh, M.; M.-Lobenhoffer, J.; Schmidt, H.; Wolf, G.; Horn, T. F. W.

Free Radical. Res., 2005, 39, 535.

38. Togna, G. I.; Togna, A.R.; Franconi, M.; Marra, C.; Guiso, M. J. Nutr. 2003, 133,

2532; Bianco, A.; Coccioli, M. G.; Marra, C. Food Chem. 2001, 77, 405.

39. Bianchi, D. A.; Blanco, N. E.; Carrillo, N.; Kaufman, T. S. J. Agric. Food Chem.,

2004, 52, 1923.; Cutler, H.G.; Majetich, G.; Tian, X.; Spearing, P. J. Agric. Food

Chem. 1997, 45, 1422.

40. Liu, J.; Birzin, E. T.; Chan, W.; Yang, Y. T.; Pai, L.-Y.; DaSilva, C. Hayes, E. C.

Mosley, R. T.; DiNinno, F.; Rohrer, S. P.; Schaeer, J. M.; Hammonda, M. L.

Bioorg. Med. Chem. Lett., 2005, 15, 715.

41. TenBrink, R. E.; Bergh, C. L.; Duncan, J. N.; Harris, D. W.; Huff, R. M.; Lahti,

R. A.; Lawson, C. F.; Lutzke, B. S.; Martin, I. J.; Rees, S. A.; Schlachter, S. K.;

Sihr, J. C.; Smith, M.W. J. Med. Chem. 1996, 39, 2435.

42. Frater, G.; Kraft, P. Helv. Chim. Acta 1999, 82, 1656.; Sprecker, M. A. US 1987,

650, 603 (Cl.252-522R,Cl 1B9/00.

162

43. Yamato, M. J. Synth. Org. Chem. Jpn., 1983, 41, 958.

44. Humber, L.G. J. Heterocycl. Chem., 1975, 12, 591.

45. McCall, J. M; McCall, R. B.; TenBrink, R.E.; Kamdar, B.V.; Humphrey, S. J.;

Sethy, V. H.; Harris, D.W.; Daenzar, C. J. Med. Chem. 1982, 25, 75.

46. Srivasta, J. N.; Chaudary, D. N. J. Org. Chem., 1967, 27, 4337; Mukhopadhyay,

D.; Chaudary, D. N. J. Indian Chem. Soc., 1963, 40, 433.

47. Mohler, D. L.; Thompson, D. W. Tetrahedron Lett., 1987, 28, 2567.

48. Bianchi, D. A.; Rua, F.; Kaufmann, T. S. Tetrahedron Lett., 2004, 45, 411.


Recommended