349
SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF ISOCOUMARINS, TRIAZOLES, THIADIAZOLES AND INDOLINONES A DISSERTATION SUBMITTED TO THE QUAID-I-AZAM UNIVERSITY ISLAMABAD IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY IN ORGANIC CHEMISTRY BY GHULAM QADEER Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan. 2008

SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

  • Upload
    others

  • View
    11

  • Download
    0

Embed Size (px)

Citation preview

Page 1: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF ISOCOUMARINS, TRIAZOLES,

THIADIAZOLES AND INDOLINONES

A DISSERTATION SUBMITTED TO THE QUAID-I-AZAM UNIVERSITY ISLAMABAD

IN PARTIAL FULFILLMENT OF THE REQUIREMENTS

FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY IN

ORGANIC CHEMISTRY

BY

GHULAM QADEER

Department of Chemistry, Quaid-i-Azam University,

Islamabad, Pakistan. 2008

Page 2: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

ISOCOUMARINS, TRIAZOLES, THIADIAZOLES AND INDOLINONES

BY

GHULAM QADEER

Department of Chemistry, Quaid-i-Azam University,

Islamabad, Pakistan. 2008

Page 3: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF
Page 4: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Allah will exalt those who believe among you,

and those who have knowledge to high ranks.

Page 5: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Saying of Holy Prophet Hazrat Muhammad

“Do you know who is the most beneficent? Allah is the

most beneficent, than of the children of the man; I am the most

beneficent and after me the most beneficent among them is the

man who acquires knowledge and spreads it, he will come on the

day of resurrection as a chief by himself”.

Page 6: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

My parents

Page 7: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

ACKNOWLEDGEMENT First of All I bow my head before “Almighty Allah” Who bestowed me the

opportunity and potential to make material contribution to the already existing

ocean of knowledge and all respects are for the Holy Prophet, Hazrat

Muhammad who enabled us to recognize our Creator.

It is a matter of great pleasure to express my obligations and sincere thanks to

my teacher and research supervisor Prof. Dr. Nasim Hasan Rama, Department

of Chemistry, Q. A. U. Islamabad, for his constant encouragement, keen interest

and thought provoking guidance through out my research work. I am thankful to

Prof. Dr S. Sakhawat shah, Chairman, Department of Chemistry, for providing

necessary research facilities. Thanks to all the teachers of organic section for

being a source of inspiration and enlightenment for me during my course work

and stay in the department.

I obliged to express my heartfelt thanks to Prof. Dr. Erik De Clercq, (Belgium) Dr.

Lieve Naesens, (Belgium), Prof. Dr. Bob Hill (Scotland), Dr. Dra. Maria Luisa

Garduño, (New Maxico), Dr. Wai Yeung-Wong (Hong Kong), Dr. Ales Ruzicka,

(Czec republic) Dr. Yong Hong Li, (China) Dr. Fan Zhijin, (China), Dr. Sauli Vuoti,

(Finland) Dr. M. Azad Malik, (U.K) James Rafetry (U.K) and Dr. Abdul Malik, H.

E. J. Research institute, University of Karachi, Pakistan for their co-operation and

generous help especially in the form of spectroscopic analysis, X-ray

crystallography and bioassay.

I ought to submit my thanks to my dear friends, who remember me in their

prayers and hearts. I wish to acknowledge the support, co-operation and

encouragement provided by Syed Jabbar Hussain Shah, Mazhar Ali Kalyar,

Shahid Ashraf, Javeed Akhter (U.K), I do remember the company of my research

fellows Dr. Muhammad Zareef, M. Arfan, Tasfeen Akhter, K. Ansar Yasin and

Page 8: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Muhammad Sher. My acknowledgement remains incomplete if I don’t mention

the help, encouragement and companionship of my lab fellows, Gul S. Khan,

Naveed Umer, Taj-ud-din, Ahsan Farid, Obaid-ur-Rehman Abid, Tariq Mehmood

Baber, Muhammad Hanif, Furrukh Iftikhar Ali, Muhammad Shahzad & Hakim

Luqman.

I am also heartily thankful to all the members of non-teaching staff of the

department especially M. Sharif Chohan, Shamas Pervaiz Qureshi, Ali Zaman,

Shamas Tabrez Qureshi, Muhammad Ilyas, M. Rashid, Muhammad Raza and

Jumma Khan, for their help during the entire period of research work.

My sincere thanks are due to my wife. Without her encouragement and

excessive generosity I would not been able to complete the task. Lastly my

special thanks are due to my parents, brothers, sisters and all of other relatives.

It is due to their prayers that I have been successful in my educational career.

May Almighty Allah shower his choicest blessings and prosperity on all those

who assisted me in any way during completion of my thesis

Thank you all,

(GHULAM QADEER)

Page 9: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

CERTIFICATE

This is to certify that this thesis submitted by Mr. GHULAM QADEER is

accepted in its present form by the Department of Chemistry, Quaid-i-Azam

University, Islamabad as satisfying the requirements for the Degree of Doctor of

Philosophy in Organic Chemistry.

SUPERVISOR ---------------------------

(Prof. Dr. Nasim Hasan Rama)

Departmet of Chemistry,

Quaid-i-Azam University,

Islamabad

CHAIRMAN ---------------------------------

(Prof. Dr. S. Sakhawat shah)

Department of Chemistry,

Quaid-i-Azam University,

Islamabad

Date:

Page 10: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

ABSTRACT

The work presented in this thesis consists of the synthesis, characterization, and

biological screening of heterocyclic compounds. For convenience, the work has

been divided into two parts, part one is related to the compounds containing

oxygen atom in the heterocyclic ring whereas part two refers to the compounds

containing nitrogen and/ or sulphur atoms in the heterocyclic ring.

Part one of this thesis is related to the synthesis, characterization and biological

activity of some heterocyclic compounds containing oxygen in the ring. These

compounds include some naturally and unnaturally occurring substituted

isocoumarins and 3,4-dihydroisocoumarins. The synthesis of naturally occurring

isocoumarin Thunberginol B is reported, in which 3,5-dimethoxyhomophthalic

acid is a key intermediate for the synthesis of Thunberginol B. It was

synthesized efficiently in five steps from 3,5-dimethoxybenzaldehyde via a series

of reactions including synthesis of 3,5-dimethoxycinnamic acid, 3-(3′,5′-

dimethoxyphenyl)propionic acid, cyclization of 3-(3′,5′-dimethoxyphenyl)propionic

acid to 5,7-dimethoxy-1-indanone and oxidative decomposition of methyl 2-

hydroxy-2-(5,7-dimethyoxy-1-oxo-1H-inden-2(3H)-ylidene)acetate to 3,5-

dimethoxyhomophthalic acid. 3,4-Dimethoxybenzoylchloride was prepared

from 3,4-dimethoxybenzoic acid on reaction with thionyl chloride which on

condensation with 3,5-dimethoxyhomophthalic acid afforded 3-(3',4'-

dimethoxyphenyl)-6,8-dimethoxyisocoumarin. Complete demethylation of 3-(3',4'-

dimethoxyphenyl)-6,8-dimethoxyisocoumarin with hydrobromic acid in acetic acid

gave 3-(3',4'-dihydroxyphenyl)-6,8-dihydroxyisocoumarin (Thunberginol B). In

addition to above, some unnaturally occurring halogenated isocoumarins and

their 3,4-dihydrodrivatives were also synthesized. The difluorophenyl- and

dichlorophenylisocoumarins by condensation of homophthalic acid with an

appropriate acid chloride. Alkaline hydrolysis of the isocoumarins yielded

corresponding keto-acids, which on reduction give the corresponding racemic

Page 11: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

hydroxy-acids. 3,4-Dihydroisocoumarins were obtained from these racemic

hydroxy-acids by cyclodehydration using acetic anhydride.

All the synthesised compounds were identified using their IR, 1H NMR and mass

spectral data. In many cases 13C NMR and elemental analysis data were

employed to support the characterization. In each case, a plasible mass

fragmentation pattern is suggested. The synthesized compounds were screened

for their antifungal, antibacterial, herbicidal, insecticidal, fungicidal, anti-

metastatic, brine shrimp lethality, antioxidant, anti -inflammatory, antiviral, anti-

HIV, anti-HBV and anticancer activities and in some cases, very fascinating

results were obtained which were then published in different international

journals. These synthetic schemes have tremendous potential for further

synthesis of novel biological active compounds.

Part two of the thesis describes the synthesis and biological screening of some,

hitherto unreported, isatin derivatives (Indolinones), 1,4-disubstituted

thiosemicarbazides and their related 2,5-disubstituted-1,3,4-thiadiazoles and 4,5-

disubstituted-3H-1,2,4-triazole-3-thiones. Indolinones were formed by the direct

condensation of hydrazides with halogenated isatins. Triazoles were formed by

intramolecular dehydrative cyclization of thiosemicarbazides in basic media while

thiadiazoles were formed in acidic media, which is an intermediate during the

synthesis of various heterocyclic compounds. Thiosemicarbazides were formed

by aldol type condensation reaction of acid hydrazides and isothiocyanates.

Isothiocyanates were formed by the reaction of anilines with carbon disulphide in

ammonium hydroxide solution to yield ammonium dithiocarbamate, an

intermediate which on oxidation with lead nitrate yield isothiocyanate. Acid

hydrazides were formed by the reaction of esters of carboxylic acid with

hydrazine hydrate and esters were formed by refluxing carboxylic acid in

methanol in catalytic amount of carboxylic acids. As a result of these synthetic

schemes, thirty new indolinones, ten disubstituted-1,3,4-thiadiazoles and twenty

disubstituted 1,2,4-triazole-3-thiones were synthesized. The characterization of

Page 12: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

these synthesized compounds was carried out by IR, 1H NMR, 13C NMR,

elemental analysis, Mass spectral data and XRD analysis. The synthesized

compounds were screened for their antifungal, antibacterial, herbicidal,

insecticidal, fungicidal, plant growth regulatory activity and antiviral activities.

Page 13: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

CONTENTS Acknowledgement I

Abstract III

Part one CHAPTER 1 INTRODUCTION

1.1 Nomenclature and structural types 1

1.2 Physical properties 5

1.3 Biological activities 5

1.4 Behavior towards human beings 12 1.5 Biosynthesis 14

1.6 Synthesis of isocoumarins and 3,4-dihydro- 18

isocoumarins

1.6.1 Synthesis involving metals 19

1.6.2 Oxidation of Indenes, Indanones and Indones 24

1.6.3 Oxidation of isochromans 26

1.6.4 Aldol type condensation between homophthalic 27

acids, esters or anhydrides and carbonyl compounds

1.6.5 Synthesis of isocoumarins via iodocyclization 32

1.6.6 Synthesis of isocoumarin by the use of new 33

technology

1.7 Interconverision of isocoumarins and 3,4-dihydro- 34

isocumarins 1.7.1 Conversion of 3,4-dihydroisocoumarins to 34

isocoumarins 1.7.2 Conversion of isocoumarins to 3,4-dihydro- 35

isocoumarins

1.8 Reactions of isocoumarins and 3,4-dihydtoisocoumarins 36

1.8.1 Hydrolysis 36

Page 14: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

1.8.2 Reaction with ammonia and amines 36

1.8.3 Reaction with phosphorus pentasulphide 37

1.8.4 Nitration 38

1.8.5 Reaction with Grignard reagents 38

1.8.6 Oxidation 39

1.8.7 Reduction 39

1.9 Plan of work 39

1.9.1 Naturally occurring isocoumarin 40

1.9.2 Unnaturally occurring isocoumarins and 3,4- 40

dihydroisocoumarins

CHAPTER 2 SYNTHESIS OF NATRUAL ISOCOUMARIN 6.1 Synthesis of Naturally Occurring Thunberginol B 41

2.1.1 Introduction 41 2.1.2 Plan of work 42 2.1.3 Results and discussions 43 2.1.4 Experimental 56

CHAPTER 3 SYNTHESIS OF UNNATRUAL ISOCOUMARINS

AND 3,4-DIHYDROISOCOUMARINS 3.1. Synthesis of Dihalophenylisocouamrins 62

3.1.1 Introduction 62

3.1.2 Plan of Work 63

3.1.3 Results and discussion 63

3.1.4 Experimental 75

3.2. Synthesis of 3H-Furo[3,4-c]-isochromene-1,5-dione 85

(an unusal isocoumarin) and 3-(3′,4′,5′-trimethoxyphenyl)

isocoumarin 3.2.1 Synthetic scheme 85

Page 15: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

3.2.2 Results and Discussion 86

3.2.3 Experimental 96

CHAPTER 4 BIOLOGICAL ACTIVITIES

4.1. Antioxidant Studies 98 4.2. Anti-inflammatory Studies 102

4.3. Herbicide studies 104 4.4. Fungicide studies 108

4.5. Insecticide Studies 111

4.6. Antifungal studies 112

4.7. Antibacterial studies 116

4.8. Brine shrimp lethality (Artemia salina) studies 118 4.9. Antiviral studies against different Cel lCulture 120 4.10. Anti-HIV Studies 124

4.11. Anti-HBV studies 127 4.12. Anti-cancer studies 129

4.13. Antimetastatic studies 131

REFERENCES (PART ONE) 133

Part two CHAPTER 5 INTRODUCTION 5.1 1,2,4-Triazole 146

5.2 Chemistry of 1,2,4-triazole 147

5.2.1 Aromaticity and stability 147

5.2.2 Amphoteric nature 148

5.2.3 Tautomerism in triazoles 148

Page 16: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

5.3 Spectroscopy of 1,2,4-triazoles 150

5.3.1 Ultraviolet spectroscopy 150

5.3.2 Infrared spectroscopy 151

5.3.3 NMR and mass spectrometry 151

5.4 Applications and biological activities 152

5.4.1 Agricultural applications 152

5.4.2 Pharmacological applications 153

5.4.3 Industrial applications 154

5.5 Synthetic approaches towards 1,2,4-triazoles 155

5.5.1 From semicarbazides 155

5.5.2 From triazine 156

5.5.3 From thiosemicarbazides 156

5.5.4 From benzalsemicarbazones with ferric chloride 157

5.5.5 From carboxylic acid hydrazides 158

5.5.6 From 1,3,4-oxadiazol-5-thiones 158

5.5.7 From thiosemicarbazides and carbonyl compounds 159

5.5.8 From thiocarbohydrazides and carbohydrazides 159

5.5.9 From thiosemicarbazides with benzoyl chloride 160

5.5.10 From phenylthiosemicarbazide with ethylphenylimidate

hydrochloride 160

5.5.11 From condensation of a nitrile and a hydrazide 160

5.5.12 From isothiocyanates 161

5.5.13 From aromatic nitriles 161

5.5.14 Solid phase synthesis of triazoles 162

5.5.15 Synthesis of 1,2,4-triazoles under microwave irradiation 163

5.6 1,3,4-Thiadiazoles 165

5.7 Applications 166 5.8 Synthetic approaches towards 1,3,4-thiadiazoles 170

5.8.1 From thiosemicarbazides 170

5.8.2 From diacylhydrazides 171

5.8.3 From dithiocarbazinic acid derivatives 172

Page 17: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

5.8.4 From Fluorous Lawesson’s reagent 172

5.9 Isatin derivatives-Indolinones 173 5.9.1 Isatin 173

5.9.2 Indolinones 174

5.9.3 Synthetic approaches towards indolinones 175

5.10 Plan of work 177 CHAPTER 6 RESULTS AND DISCUSSION

6.1 Synthesis of methyl / ethyl esters 180 6.1.1 Crystal structure of Ethyl 2-(3-methoxyphenyl)acetate 182

6.1.2 Crystal structure of Methyl 2,6-dimethoxybenzoate 185

6.2 Synthesis of hydrazides 187 6.2.1 Crystal structure of 3,5-difluorobenzohydrazide 189

6.2.2 Crystal structure of 2,6-dimethoxybenzohydrazide 192

6.2.3 Crystal structure of 3,4-dimethoxybenzohydrazide 193

6.2.4 Crystal structure of 2-(2′,4′-dichlorophenylsulfanyl)-

acetohydrazide 196

6.2.5 Crystal structure of (E)-3-(4′-methoxyphenyl)acrylo-

hydrazide 198

6.2.6 Crystal structure of 3-(3′,4′,5′-trimethoxyphenyl)-

propanehydrazide 200

6.2.7 Crystal structure of 3-(4′-methoxyphenyl)-

propanohydrazide 203

6.3 Synthesis of isothiocyanates 205

6.4 Synthesis of thiosemicarbazides 207 6.4.1 Crystal structure of 1-(3,5-Dimethoxybenzoyl)-4-(2-

methoxyphenyl) thiosemicarbazide 212

6.4.2 Crystal structure of 1-[2-(2,4-Dichlorophenoxy)-

acetyl]-4-cyclohexylthiosemicarbazide 214

6.4.3 Crystal structure of 1-[3-(4-methoxyphenyl)

Page 18: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

propanoyl]-4-(2- methoxyphenyl)thiosemicarbazide 216

6.4.4 Crystal structure of 1-(3,5-Difluorobenzoyl)-4-

Cyclohexylthiosemicarbazide 217

6.5 Synthesis of substituted 1,2,4-triazol-3-thiones 219

6.5.1 Crystal structure of 4-(2-methoxyphenyl)-5-(3,5-

dimethoxyphenyl)-2H-1,2,4-triazole-3(4H)-thione 224

6.5.2 Crystal Structure of 5-(3,4,5-trimethoxyphenylethyl)-

4-(2-methoxyphenyl)-2H-1,2,4-triazole-3(4H)-thione 227

6.5.3 Crystal Structure of 3-(4-Methoxyphenethyl)-4-(2-

methoxyphenyl)-1H-1,2,4-triazole-5(4H)-thione 229

6.5.4 Crystal Structure of 3-(4-bromophenoxymethyl)-

4-(4-methoxyphenyl)-1H-1,2,4-triazole-5(4H)-thione 231

6.5.5 Crystal Structure of 3-(2,4-dichlorophenoxymethyl)-

4-(4-methoxyphenyl)-1H-1,2,4-triazole-5(4H)-thione 233

6.6 Synthesis of 1,3,4-thiadiazoles 235 6.7 Synthesis of indolinones 239

6.7.1 Crystal Structure of (Z)-N'-(5-chloro-2-oxoindolin-

3-ylidene)-3,5 difluorobenzohydrazide 243

CHAPTER 7 EXPERIMENTAL

7.1 Substrates and reagents 245 7.2 Purification of solvents 245 7.3 Instrumentation 246 7.4 General procedure for the synthesis of esters 247 7.5 General procedure for the synthesis of hydrazides 250 7.6 General procedure for the synthesis of isothiocyanate 256 7.7 General procedure for the synthesis of

Thiosemicarbazides 257 7.8 General procedure for the synthesis of 1, 2, 4-

triazol-3-thiones 267 7.9 General procedure for the Synthesis of 1,3,4-

Page 19: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Thiadiazoles 276 7.10 General procedure for the synthesis of Indolinones 280

CHAPTER 8 BIOLOGICAL ACTIVITIES

8.1 Herbicide studies 294 8.2 Fungicide studies 296 8.3 Insecticide studies 298 8.4 Plant growth regulating studies 300 8.5 Antiviral studies against different cell cultures 302 8.6 Antifungal studies 310 8.7 Antibacterial studies 314

REFERENCES (PART TWO) 316

Page 20: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Chapter-1 INTRODUCTION

Isocoumarins and 3,4-dihydroisocoumarins are the secondary metabolites1.

These are found in wide varieties 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, and

Streptomyces etc. A number of them are constituents of a few higher plant

families e.g. Bignoniaceae, Compositae, Leguminoseae, Myricaceae,

Saxifragaceae, Myristicaceae and Liliaceae families. A number of reviews have

been published about isocoumarins. These include the review by Barry2 (1964),

Turner and Aldridge3 (1983), Yamato4 (1983), Hill5 (1986), Napolitano6 (1997)

and Bin7 et al. (2000) on isocoumarins and 3,4-dihydroisocoumarins.

1.1 Nomenclature and structural types

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

are isomeric to coumarins (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

R1

R2R3R4

R5R6

12

3456

78

4a

8a

O

O

R1

R2R3R4

R5R6

1 2

3

456

7 8

4a

8aO O

(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

1

Page 21: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

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), 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. Example 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-R6=H) had 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-R6 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

2

Page 22: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

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, R2=COOH) and (6, R1=CHO, R2=H) are

called as 7-carboxymellein and 5-formylmellein respectively.

O

OOH

O

OOH

O

OOH

OR1

R2

(4) (5) (6)

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

ustic acid (8) and its derivatives exist in tautomeric equilibrium between their

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

O

OH

C15H31

O

OOH

C15H31HO

(7a) (7b)

COOH

HOHO

O

OOH

HO

(8b)

OH

O

OH(8a)

COOH

HOOHO OHO

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

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

3

Page 23: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

number of variously substituted isocarbostyrils10 and tetrahydroisoquinolinones

(10), which can also exist as its other tautomer, have been prepared.

NHR

O

NR

OH

NHR

O(9a) (9b) (10)

Sulphur analogues have also been known since times 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

S

O

S

S

(11) (12) (13)

OR

In 1980, a three-step synthesis of 2-seleno- and 2-telluroisocoumarins

was reported12. Regiospesific nucleophilic β-addition of methaneselenolate or

methanetellurolate 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.

`

OR

Y

O

X

O

14a) R = C2H5, Y = Se or Te14b) R = H, Y = Se or Te

15) X = Se, m.p. 80°C16) X = Te, m.p. 83° C

4

Page 24: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

1.2 Physical properties

Isocoumarins are usually solid crystalline compounds having melting points

ranging from 49-50°C (trans-artemidin) to 350°C (alternariol). Some

isocoumarins like 3-pentylisocoumarin and 3-propylisocoumarin are oils. It is

observed that melting points of isocoumarins are invariably higher than those of

corresponding dihydroisocoumarins.

1.3 Biological activities

In 1964 when the review by R. D. Barry2 appeared there were only a few

valid reports of the biological activities of isocoumarins. A literature survey of the

later period shows that isocoumarins display a wide range of biological activities.

A large number of 3-phenylisocoumarin13 has been tested for various

pharmacological activities. Some of these are useful sweeteners, anti-corrosives,

fluorescent agents and laxatives, whereas others possess anti-inflammatory,

anti-allergic, anti-malarial activities and have proved to be useful in the treatment

of asthma14-15.

Mellein (17) has been found in several insects. The defensive secretion of

termites16, Australian onerine ants17, the mandibular gland secretion of

Camponotus herculeanus, C. lighiperda and C. pensylvanicus18 (carpenter ants)

and the male hair pencil of the oriental fruit moth19, all contain mellein. Mellein

and its dihydroderivatives20 are found in the defensive secretions of tenebrionid

beetle, Apsena pubescencs.

O

OOH

(17) Many fungal isocoumarins exhibit antifungal activities21 particularly

oospolactone22 (18), chladosporin23 (19), 6-methoxymellein24 (20), 3-phenyl-3,4-

5

Page 25: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

dihydroisocoumarin-4-carboxylic acid25 (21) and 3-phenyl-4-(hydroxyacetyl)-3,4-

dihydroisocoumarin (22).

Coriandrin (23), one of the two naturally occurring furoisocoumarins

known to date, was isolated in 1988 from dry coriander leaves26. In addition to

the expected psoralen activity, it shows in vitro anti-HIV activity27.

O

OOH

O

O

(20)

OH

OHO O

O

OOH

(18) (19)

O

COOH

O

O

O

OHO

(21) (22) (23)

O

O

O

6-[2-Chloro-4-(trifluoromethyl)phenoxy]-3,4-dihydroisocoumarin (24) has

shown herbicidal activity28 and its application of 1kg/ha almost totally controlled

Schinochloa crus-galli, Sinapis alba and other weeds.

Twenty isocoumarins derivatives29 were tested for biological activities

towards rice, radish, barnyard grass and A. niger. At 100 ppm, 4-carboxy-6-

chloro- (25, R=R1=H, R2=6-Cl), 4-carboxy-7-chloro- (25, R=R1=H, R2=7-Cl), and

6-chloro-4-ethoxycarbonyl-3-methylisocoumarin (25, R=Me, R1=Et, R2=6-Cl), were phytotoxic to radish and rice plants while 4-ethoxycarbonyl-6,7-dimethoxy-

3-methylisocoumarin (25, R=Me, R1=Et, R2=6,7-(OMe)2), was phytotoxic to

radish. 3-Methyl-, 3,7-dimethyl- and 6-methoxy-3-methylisocoumarins inhibited

the growth of A. niger.

6

Page 26: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O(24)

OCl

F3CO

RCOOR1

O

R2

(25)

Several isocoumarins30 (26) (R=H, alkyl, alkenyl, alkoxy, nitro) are useful

as antihypertensives, antiarrhythmics and β-sympatholytics. These were

prepared starting from 3-hydroxyhomophthalic acid. Antiarrhythmic activity of

(26) (R=H) was comparable to that of pindolol (standard).

Isocoumarins 27(a-b) with different substituents, isolated from the fungus

Ceratocysistis fimbriata coffea, were found to have toxic activity on coffee tree

leaves31 and horse radish peroxidase. Compound (27b) also exhibits antiviral

activity as well as a distinct inhibiting activity on 3α-hydroxysteroid

dehydrogenase (3α -HSD) 32.

Cytogenin (28) shows antitumour activity33 against Ehrlich carcinoma at

6.3 to 100mg/ Kg/ day.

(26)

O

O

RRR

R

ROHO

NH

O O

O O

R1R2

OH OH

OOH

(28)27a) R1 = CH3, R2 = OCH327b) R1 = CH3, R2 = OH

Some derivatives34 of isocoumarin and thioisocoumarin (29) (R=H, OH,

NO2, NH2, halo, alkyl; Y=H, halo, OCH3, CF3; X=O, S; Z=H, halo, C1-6 alkyl,

7

Page 27: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

alkylphenyl etc.) are serine protease inhibitors and useful in treatment of

emphysema.

Five isocoumarins35 (-)-S-5-methyl, (-)-S-5-carboxy-, (-)-5-hydroxyethyl-

mellein, cis- and trans-4-hydroxy-5-methylmellein (30) isolated from pathogenic

fungus of apple canker, Valsa ceratosperma showed phytotoxicity in apple

shoots and the lettuce seedlings.

XR

Z

O

(30)

Y

O

O

RR

OH

(29)

3-Alkoxy-7-amino-4-chloroisocoumarin derivatives (31) were synthesized

as new beta-amyloid peptide production inhibitors and found very active against

various classes of proteases36-38.

The isocoumarinyl penicillin derivatives39 (32) were quite effective

bacteriocides at 3.15-100 mg/mL.

O

O

OR

H2N

(32)

O

O

O

NH

N

S

ONaO

O

(31)

Cl

A new isocoumarin 2-(8-hydroxy-6-methoxy-1-oxo-1H-2-benzopyran-3-

yl)propionic acid (NM-3)40-41 (33), which is an analogue of the natural product

cytogenin, induces the lethality of human carcinoma cells by generation of

reactive oxygen species42 and inhibits angiogenesis43-44. It increases the

antitumour effects of radiotherapy with toxicity45 and Potentiates

Dexamethasone-Induced Apoptosis of Human Multiple Myeloma Cells46. It also

8

Page 28: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

increases the antitumor effects of various existing chemotherapeutic drugs in

breast and prostate tumor model systems, as measured by TGI47.

Dihydroisocoumarin PF1223 (34) from the culture of Neosartorya

quadricincta inhibit the specific binding of the non competitive antagonist [3H]

EBOB to housefly head membranes. This compound at 2.2 µM inhibited [3H]

EBOB binding by 65%48.

(33)

O

OH

O

O

O

OH O

OOH

O

O

OH

O

OH

(34)

Phytochemical investigation of the aqueous extract of the roots of

Agrimonia pilosa Ledeb (Rosaceae), as guided by hepatoprotective activity in

vitro, furnished two isocoumarins, agrimonolide (35a) and agrimonolide 6-O-β-D-

glucoside (35b). Compound (35a) showed hepatoprotective effects on both

tacrine-induced cytotoxicity in human liver-derived Hep G2 cells and tert-butyl

hydroperoxide-induced cytotoxicity in rat primary hepatocytes with EC50 values

of 88.2 ± 2.8 and 37.7 ± 1.6 µM, respectively49.

Reticulol (36), produced from a strain of Streptoverticillium, was found to

be active against Topo I mediated DNA relaxation in vitro50. The treatment with

45 µM reticulol afforded inhibitory activity, but with 0.45 and 4.5 µM reticulol,

relaxation of DNA was not significantly reduced. The potency of 45 µM reticulol

in inhibiting relaxation was almost the same as that of 0.1 mM camptothecin

(molecular weight 348.34). Consequently, reticulol exhibited Topo I-inhibitory

efficacy similar to a positive control, camptothecin. This result demonstrated that

reticulol blocked the relaxation of DNA with the formation of supercoiled DNA by

inducing the inactivation of Topo I.

9

Page 29: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

OOH

RO

O

35a) R = H35b) R = Glc

O

HO

OOH O

(36)

Seven new naturally occurring 3-butylisocoumarins (37) were isolated and

identified from the lipophilic extracts of aerial as well as underground parts of

Asteraceae-Anthemideae51. The antifungal activities of all naturally occurring

derivatives were determined in a germ-tube inhibition test against a susceptible

strain of rice blast fungus Pyricularia grisea. The 3-butyl side chain is

prerequisite for high activity.

Eleven new guanidino-, amino alkoxy- and isothiureido alkoxy substituted

isocoumarins52 (38) are potent mechanism based inhibitors for blood coagulation

serine proteases and other trypsin-like enzymes. In many cases, the inhibited

enzymes are very stable. These isocoumarins are effective anticoagulants in

human plasma.

A substituted isocoumarin53 (39) had a dose dependent reproducible

anticoagulant effect in rabbit. Its effect however ceased shortly after stopping

administration, indicating that its half-life is very short.

(37)

O

O O

OH2NHN

Cl

O

O

OH2N

ClS NH2

NH

(38) (39)

10

Page 30: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Isocoumarins substituted with chloro and basic groups such as guanidino

and isothiureido alkoxy are inhibitors of complement serine proteases. 3,4-

Dichloroisocoumarins are general serine protease inhibitor and 4-chloro-3-(3-

isothiureiodopropxy)isocoumarin (40) is found to be the best inhibitor54 .

Several 6,8-dihydroxyisocoumarins and 6,8-dihydroxy-3,4-dihydro-

isocoumarins are the metabolites of phytopathogenic fungi. The 3-methyl-3-

hydroxy-6,8-dihydroxy-3,4-dihydroisocoumarins (41) and 3,4,6,8-tetrahydroxy-3-

methyl-3,4-dihydroisocoumarin (42) implicated as possible phytotoxins produced

by ceratocystis ulmi, the fungus responsible for Dutch Elm disease55-56. The

dihydroisocoumarin (42) causes lesion on the leaves of pear trees and inhibit the

growth of rice seedlings57.

O

O

Cl

Oisothiuredine

(40)

O

OH

HO OH

O

O

OH

HO OH

O

OH

(41) (42)

8-Hydroxy-3,4-dihydroisocoumarins have been found to constitute a new

class of ant pheromone58. The insects use trail pheromones for making trail

between the feeding place and the nest. Mellein and other 8-

hydroxydihydroisocoumarins 43(a-d) and 3-ethyl-7-methyl-8-hydroxy-3,4-

dihydroisocoumarin (44) have been isolated as trail pheromones in the hindgut

of ants of various species of the genera Formica and Lasius found in Germany.

11

Page 31: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

OH

R1

O

O

OH O

R

R2

43a) R = R1 = R2 = H 43b) R =R1 = Me, R2 = H43c) R1 = R2 = Me, R = H 43d) R = R2 = Me, R1 = H

(44)

Sclerin (45) and sclerotinins A (46) and sclerotinins B (47), metabolites of

sclerotinia sclerotiorum have plant growth regularity effect59-60.

O

OH

O

O

O

OH O

HOR

46) R = CH347) R = H

(45)

The mode of action of these isocoumarins in phytopathogenic organisms

is not clear although they are observed to build up in the trees and plants after

infection by the fungi61.

1.4 Behaviour towards human beings

Some isocoumarins have beneficial activities while others are harmful

towards human beings. Some show diuretic and antihypertensive activities62 (48) (R=Phenyl, 3,4-dichlorophenyl, 4-methoxyphenyl, R1=H, methyl, NR2R3=NMe2,

N-diallyl, 4-methylpiperazino, morpholino , etc.). Some have been used in the

treatment of lymphedema63. The antitumour activity of duclauxin (49) has been

demonstrated64. Analogues of bactobolin A (50) have antileukemic activity65

whereas bactobolin A and its related metabolites are active against bacteria and

viruses66. Among the most important isocoumarins, the AI-77s (51), endowed of

12

Page 32: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

gastroprotective properties67-68 but free of effects on the central nervous system,

Phyllodulcin, a lead compound in the discovery of novel low calorie sweeteners,

coriandrin, active against HIV69. Agrimonolide possesses strong anthelmintic

activity70. The observations of scanning electron microscope have shown that

anthelmintic effects of agrimonolide are due to its destructive effects on the body

wall of tapeworms. A number of isocoumarin-3-carboxylic acids71 (52) (R, R1, R2

= H, methyl, methoxy, bromo, etc.) were active in the passive cutaneous

anaphylaxis test.

O

OOAcO

HO

O

OH

O

O

O

O

N R3

R R1 R2

O

O

NH

OH

O

OOH

O

O

COOH

O

R2

R1

R

O

OOH

HOCHCl2

HNH

OHNH2

O

(48)

(49)

(50)

(51) (52)

Some isocoumarins and dihydroisocoumarins are harmful to man, like

ochratoxin A (53) and ochratoxin B (54) which are nephratoxic and hepatotoxic

metabolites of several Aspergillus and Penicillium species72-73. Ochratoxin A (53)

inhibits protein synthesis74. Oosponol inhibits dopamine β-hydroxylase and

causes severe skin rash, bronchitis and pneumonia75 and reticulol (36) inhibits

cyclic AMPase76.

O

OH O

HN

COOHO

O

OH O

HN

COOHO

Cl

(53) (54)

13

Page 33: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

1.5 Biosynthesis

J. N. Collie (1907) made the first suggestion of the biosynthesis of certain

classes of aromatic compounds, by the head to tail condensation of acetate

units. These considerations were based upon the reactivity of synthetic linear β-

polyketones (55; n ≥2) which underwent aldol type condensations to form

aromatic phenolic compounds.

R

OO

n

(55) Biogenetic definitions of the aromatic polyketides are due to the work of

Birch (1951), by whom a detailed theoretical analysis of carbon skeletons and

oxygenation patterns of known compounds was first combined with an extensive

series of tracer incorporation experiments. The elaboration of the β-ketide chain

in metabolites derived from poly-β-ketide precursors, normally proceeds by

condensation of terminal unit of acetyl Co-enzyme A with chain building units of

malonyl Co-enzyme A (Scheme 1.1).

SCoA

O

HSCoA + CO2

+ HO SEnz

O O

SEnz

O O

Scheme 1.1 The orsellinic acid may be derived from a β-tetraketone, which might undergo

the aldol or claisen type condensation (Scheme 1.2).

SEnz

O OO O

OHHO

OH

OHHO

OH

O

O

a

b

Scheme 1.2

14

Page 34: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

T. Money and F. W. Comer et al.77 using pyrones as masked β-

polycarbonyls provided the experimental verification of the acetate-malonate

pathway for the synthesis of isocoumarins. Thus hydrolysis of trypyrone (57)

which is a protected β-pentaketide (56) afforded eight crystalline compounds, six

of which were obtained by internal aldol condensations of (57) at positions 2 and

7 (Scheme 1.3). The products (58) and (59) appeared due to prior degradation

of (56 or 57). The remaining products represented conversion of the intact C10

chain and are variants of the 2,7-aldol condensation of (57). The most significant

of these are 6,8-dihydroxy-3-methylisocoumarin (60, R = H) and 7-

carbomethoxy-6,8-dihydroxy-3-methylisocoumarin (62, R = COOCH3). The

structure of (62) was confirmed by spectroscopy and its conversion to dimethyl

ester (63, R=COOCH3) which is another product of the reaction. The methyl

ether (64) has been isolated from Endothia parasitica, the lactol viz. 3,4-dihydro-

3,6-dimethoxy-8-hydroxy-3-methylisocoumarin (65) has also been isolated.

O OO O

OR

O

COORCOOR2378

(56)

Scheme 1.3

O

O

O

O

O

OHO

23

78

OO

O

OH

O

O

COOCH3

O

O

OH

O

HO OH

OO

OH

HO

O O

O

HO

OH OR COOCH3

O

HO

OHR

60) R = H62) R = COOCH3

61) R = H63) R = COOCH3

(57)

(58)

(59)

(65)

(64)

15

Page 35: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

This biogenetic type synthesis of isocoumarins and related compounds

confirmed the acetate-malonate pathway for biosynthesis of such compounds. 14C Labeled malonate in these metabolites yields a product in which each of the

chain building unit carries a label but the terminal unit of the chain is inactive. On

the basis of structure analysis and tracer work many fungal metabolites appear

to be derived biogenetically from the acetate and polymalonate pathway78. For

example mellein is formed from acetate and malonate as given in (Scheme 1.4).

O

O

COOH

CH3

OO

O

HO

COOHOOCH3COOH 4H2C

COOH

COOH

OCOOH

OHO

O

O

OH

Scheme 1.4

The early reduction of two carbonyl groups in the polyketide chain

followed by the loss of oxygen function at C-6 and then aldol type condensation

result in the aromatization, to give mellein. Loss of the oxygen function at C-6 of

an isocoumarins is quite common but loss of the hydroxyl group at C-8 never

occurs in those isocoumarins derived from acetate presumably a consequence

of the cyclization mechanism79.

In 1970 Y. Suzuki80 isolated fusamarin (66) from a strain of fusarium sp.

first synthesis was completed in 1978 under the supervision of Prof. W. B.

Whalley 81. In the biosynthesis of fusamarin the main chain may be derived from

poly-β-ketide pathway and is biosynthesised from seven acetate units.

16

Page 36: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

OH

HOH

O

(66) The butyl chain at C-5 is supposed to be derived from isopentyl

pyrophosphate in which one carbon atom is missing presumably may involve an

electrophilic attack on the poly-β-ketide chain. This would be a unique example

of the deviation from the "biogenetic isoprene rule". 6,8-Dihydroxy-3-undecyl-3,4-

dihydroisocoumarin82 (67) isolated from Ononis natrix and peniolactol83 (7a, 7b)

isolated from Peniophora sanguinea Bres belong to poly-β-ketide pathway and

can be biosynthesized from ten and twelve acetate units and their synthesis

have been completed under the supervision of Prof. N. H. Rama84-85.

O

O

OH

OHHO

O

OH

HO

COOH

(7a)

(7b)

Biosynthesized from twelve acetate units

O

O

OH

HO

(67)

Biosynthesized from ten acetate units

17

Page 37: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

The biosynthesis of plant derived isocoumarins has been studied to a

lesser extent than fungal isocoumarins. Phyllodulcin (69), the sweet principle of

Hydrangea macrophylla and hydrangenol (68), has been shown to be formed

from phenylalanine via cinnamic acid and p-coumaric acid with the addition of

three acetate units86-89 (Scheme 1.5).

OHNH2

O

OH

O

OH

O

HO

O

O

OH

OH

O

O

O

OH

OH

(68)(69)Scheme 1.5

Bergenin (70) is derived from C-glucosylation of gallic acid and

subsequent lactone formation90 (Scheme 1.6).

Scheme 1.6

O

OO

O OHCH2OH

OHH

HO

O

OH

OHHO

HO

Glucose

(70)

1.6 Synthesis of isocoumarins and 3,4-dihydro- isocoumarins

A wide spectrum of synthetic methods have been used towards the

synthesis of isocoumarins and 3,4-dihydroisocoumarins91. A number of new

18

Page 38: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

methods 92-102 are being developed and reported each year. Some of these

methods provide the isocoumarins directly whereas others lead to the 3,4-

dihydroisocoumarins. Some of the most important, high yield methods applicable

to the synthesis of a large number of these compounds are mentioned below.

1.6.1 Synthesis Involving Metals

Literature reveals that isocoumarins and 3,4-dihydroisocoumarins have

been extensively prepared by such methods, involving metallation at specific

position like lithiation, silylation and thallation etc.

a. Lithiation Reaction

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 reviewed103-105. Summarizing the general concepts, carboxylic acid

derivatives suitable for promoting ortho lithiation106-107 are tertiary amides (4,4-

dimethyl)oxazolin-2-yl group and secondary amides. Lithiated tertiary amides are

readily and generaly ortho-lithiated using s-butyllithium and tetramethylethylene-

diamine, 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

19

Page 39: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

demonstrated by the enantioselective synthesis of the isocoumarin portion of

AI77B (71) (Scheme 1.7)108-111.

O

OOO

N

O

a,b,c

O

N

O

d,e

OH

O

O

NH2

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

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

Scheme 1.7

(71)81% 69.6% OH

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 reaction112. Good

yields have occasionally been reported though, as in the synthesis of the

allergenic principle of gingko biloba (72) (Scheme 1.8)113 and of a variety of

mellein derivatives114.

O

C12H25

OOO

LiN

O

a

O

HN

O

bO

O

a) (R)-1,2-epoxytetradecane b) OH, then neutralization CuCN(LiCl2) c) BBr3

Li

C12H25 OH

OH

C12H25

70%98%

c

_

Scheme 1.8

(72)

Lateral lithiation of (S)-4-isopropyl-2-(o-tolyl)oxazoline in diethyl ether

followed by the reaction with aldehydes in the presence of TMEDA produced the

addition products with stereoselectivities up to 84% de115. Utilization of TMEDA

20

Page 40: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

as a ligand is essential for the good selectivity. Rationale for the stereoselectivity

is proposed based on ab-initio calculation of the lateral lithio species. The major

(S,S)-products lactonized faster than the minor (S,R)-products to the

corresponding 3,4-dihydroisocoumarins under acidic conditions. Thus, (3S)-3,4-

dihydroisocoumarins were obtained in good optical purities up to 97% ee by

sequential application of these matched stereo-selective reactions (Scheme 1.9)

LiN

ON

O

RHO*

O

O

R*

RCHO H3O+

Scheme 1.9

b. Silylation Method

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

benzamides, which generates carbanions suitable for additions to aldehydes116.

2-(Trimethylsilylmethyl)benzoyl chlorides also undergo desilylation and addition

to aldehydes to give dihydroisocoumarins through a concerted mechanism

involving ortho-quinodimethanes rather than carbanions as reactive

intermediates (Scheme 1.10)117.

Si(CH3)3Cl

OOO

OH

O

a

O

OH

O

bO

Ar

O

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

O

cSi(CH3)3

50-53%

Scheme 1.10

OO

To this class of reactive intermediates belong 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 1.11)118.

21

Page 41: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O O

a,b

O

CN

O

cO

Ph

O

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

OOHO

CNCN

Scheme 1.11

c. Thallation-olefination of Arenes

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

reaction119, 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 1.12).

Tl

COOH

O

O

RRH2C=CH2

PdCl2

Scheme 1.12

d. Palladium catalyzed method

Aryl iodides with a nucleophilic substituent at the ortho position react with

1,2-dienes in the presence of a palladium catalyst120 and a chiral bisoxazoline

ligand to afford isocoumarin (73) in good yield and with 46-86% enantiomeric

excess (Scheme 1.13).

22

Page 42: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

I

COOHO

O

n-C8H17

+ n-C8H17-CH=C=CH2

5mol% Pd(dba)25mol% Ligand

1.2 equiv Ag3PO4

Scheme 1.13

(73)

e. Iridium catalyzed method

Two new cyclizations of ketoaldehydes121 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 (74a-b) in good yields (Scheme 1.14).

R

O

O

R

OH

O

R

O

O+

OIr

HN

PhPh

(5mol%)cooxidantbast, rt, 16 h

R= CH3R= Ph

R

O

O

R

O

O+

Ir cat (5mol%)

t-BuOH, reflux

R

O

O

74a) R= CH3, 70%74b) R= Ph, 45%

28%47%

Scheme 1.14

f. 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 catalyst122 and

oxidant respectively to produce the corresponding isocoumarin derivatives. The

copper salt can be reduced to a catalytic quantity under air (Scheme 1.15).

23

Page 43: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

COOHO

OScheme 1.15

H+ R R

Rh-Cat.

Cu-Salt

RR

g. Mercury catalyzed method

Sulphuric acid–catalyzed chloralhydrate condensation with different m-

substituted benzoic acids formed trichlorophthalides, from which Zn+AcOH

reduction afforded various dichloro derivatives. These derivatives on treatment

with alkaline Hg(OAc)2 + I2 furnished different substituted isocoumarins123

(Scheme 1.16).

COOHRRO

O

CCl3Zn-AcOH

Cl

Cl RO

O

ClHg(OAc)2NaHCO3, I2

DMSO70-80°C3-4h 75a) R= OCH3

75b) R= OH75c) R= OCH2PhScheme 1.16

1.6.2 Oxidation of Indenes, Indanones and Indones

Isocoumarins and 3,4-dihydroisocoumarins have been prepared in high

yield by ozonization of indene (76) in ethyl alcohol, followed by decomposition of

the intermediate cyclic perester124 (77). Treatment of 2-

carboxyphenylacetaldehyde (78) with mineral acid (or copper powder) 125 lead to

isocoumarin and sodium borohydride give 3,4-dihydroisocoumarin (Scheme 1.17).

24

Page 44: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O3

OO

HO

OC2H5OH

OO

O

O

O

O

O

O

OH

O

ONaBH4

AlkAlk

H+

(76) (77)

(78)

Scheme 1.17 Oxidative cleavage of indanone has been used for synthesis of 6,8-

dimethoxy-3-methylisocoumarin126-127and related isocoumarins. Thus 2-

methylindan-1-one (79) was converted into silyl ether (80) which on ozonolysis

afforded the 2-hydroxyindanone (81). The latter on periodate cleavage afforded

the isocoumarin (82) presumably via the keto acid (82a). Alternatively, the

conversion of indanone (79) into enol (81) followed by ozonolysis gave the

desired isocoumarin (83) (Scheme 1.18).

O

R

OOSi(CH3)3

R

O OSi(CH3)3

R

OO

OCOCF3

R

OO

R

OOH

R

O

R

OCOOH

OO

O

Scheme 1.18

(79) (80)

(81)

(82a)

(82)

(83)

25

Page 45: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Indanone epoxide (84) prepared from indanone by epoxidation with H2O2/

(C2H5)3N in acetone was submitted to flash vacuum pyrolysis128 (FVP)

(450oC/0.1 mm).The epoxide undergoes rearrangement during FVP to afford

isocoumarin129 (85) (Scheme 1.19).

OO OO

(C2H5)3N, H2O2

OO

O

O

OO

R

O

OO O

O

FVPπ4a+π2acycloreversion

(84)

(85)Scheme 1.19

1.6.3 Oxidation of Isochromans

Oxidation of isochromans with a variety of reagents e.g. selenium dioxide,

chromium trioxide, potassium permanganate, nitric acid or air yields the

corresponding 3,4-dihydroisocoumarins (Scheme 1.20).

OR O

R

OScheme 1.20

Isochroman formed from 2-arylethanol130 is also converted to 3,4-

dihydroisocoumarin by oxidation with PCC in boiling CH2Cl2129, which give

isocoumarin (86) (R=H, 7-Me, 5-CF3, 5,6-C4H4) on treatment with NBS and Et3N

(Scheme 1.21).

26

Page 46: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OR O

R

O

OR'R

(R' = H, MEM)

a bO

R

O

c

b) PCC c) NBS, (C2H5)3N(86)

Scheme 1.21

a) TiCl4

1.6.4 Aldol-type Condensation between Homophthalic Acids, Esters or Anhydrides and Carbonyl Compounds

This type of condensation is mostly used in the synthesis of isocoumarins

and 3,4-dihydroisocoumarins. The most important methods of aldol type

condensation are discussed in four main groups.

a. Stobbe Condensation of Homophthalates with Aldehydes and Ketones

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

dihydroisocoumarins131-135. Synthesis of (dl)-agrimonolide136 provides a good

example of application of Stobbe condensation.

Thus, diethyl 3,4-dibenzyloxyhomophthalate (87) 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 (88a,

R=COOEt). Hydrolysis and decarboxylation gave 2,4-dibenzyloxy-6-[4-(4'-

methoxyphenyl)buten-1-yl]benzoic acid (88b, R=H) which on cyclization with

bromine gave the 4-bromo-3,4-dihydroisocoumarin (89). Reductive

debromination and debenzylation was simultaneously effected by adding triethyl

amine to the catalytic reduction medium to furnish the (dl)-agrimonolide (90) (Scheme 1.22)

27

Page 47: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

BzO

OBz

OMeBr

O

O

COOC2H5

COOC2H5OBz

BzO

OBz

BzO

COOH

R O

O

O

NaOH

88a) R= COOC2H5

OBz

BzO

COOH

O

Br2/CHCl3

HO

OH

OMe

O

O

H2/Pd-C/(C2H5)3N

(87)

88b) R = H (89)

(90)

Scheme 1.22

b. Claisen Condensation of Homophthalates with Formates

Diethyl homophthalate (91) condenses with methyl formate in the

presence of sodium ethoxide imparting a 66% yield of isocoumarin-4-carboxylic

acid (92). Decarboxylation with phosphoric acid furnishes isocoumarin (93)137

(Scheme 1.23).

28

Page 48: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

COOC2H5

COOC2H5

HCOOCH3

C2H5ONa O

O

COOH

H3PO4

-CO2O

O(91) (92) (93)

Scheme 1.23

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 yield138.

c. Claisen Condensations of Homophthalates with Oxalates

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

condensation between diethyl homophthalate (94) and diethyl oxalate, giving a

67% yield of triester (95). This triester loses ethanol when heated yielding diethyl

isocoumarin-3,4-dicarboxylate (96). Under different hydrolysis conditions

different products are formed. Thus heating (96) at 68-72°C for 3hr. gives ethyl

isocoumarin-3-(carboxylic acid)-4-carboxylate (97) and prolonged heating yields

isocoumarin-3-carboxylic acid (98). Boiling hydrochloric acid or heating in a

sealed tube at 180-190°C converts (96) to isocoumarin-3-carboxylic acid in 84%

yield139. These results indicate that the ester at position 3 in (96) is hydrolyzed

first, but the acid at position 4 is more easily decarboxylated (Scheme 1.24).

29

Page 49: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

COOC2H5

COOC2H5COCOOC2H5

OC2H5NaOO

OO+

O

O O

O

COOC2H5

COOC2H5

O

O

O

COOH

O

O

COOHCOOC2H5

(94)(95)

(96)

(97)

(98)

Scheme 1.24

d. Condensation of malonyl heterocycles with diphenylcarbonate

Reaction of diphenylcarbonate (100) with enolized phenylmalonyl

heterocyclic compounds as (99 a-d) yields the condensed isocoumarins140 like

(101 a-d) (Scheme 1.25).

X

OH

O X

O

O

O

C6H5O OC6H5

O

a) X= Hb) X= NHc) X= NCH3d) X= NC6H5

(99) (101)

(100)

Scheme 1.25

30

Page 50: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

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

Tirodkar and R. N. Usgaonkar141-142 carried out two or three step

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

catalysed acylation of homophtalic acids with acid chlorides or anhydrides to

give isochroman-1,3-dione (102). Treatment of (102) 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 1.26).

COOH

COOHR (R'CO)2O/Py

90-100°C RO

R'COOH

Or.t

RO

OCOR'

O

RO

R'

O

conc H2SO4

90% H2SO4

(102)Scheme 1.26

S. Nakajima et. al. synthesized various 3-arylisocoumarins (104, Ar =Ph,

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

(80%) by heating directly the homophthalic acids (103, R, R1, R2=H, OH, OMe,

Cl) with aryl or acyl chlorides at 190°C. These isocoumarins were converted into

corresponding 3,4-dihydroisocoumarins by reducing with NaBH4(Scheme 1.27).

COOH

COOH

Ar/ RCOCl190°C O

Ar/ R

O

RR1

R2

RR1

R2(103) (104)

Scheme 1.27

31

Page 51: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

A. Rose143 and later on H. Yoshikawa144 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 (PPA) or anhydrous stannic chloride e.g. 7-methyl-3-(2′-hydroxy-4′-

methylphenyl)isocoumarin (105) was obtained from 7-methylhomophthalic acid

(Scheme 1.28).

Scheme 1.28

COOH

COOHO

O

OH

(105)

PPA

anh. SnCl4

3-(2',4'-Dimethoxyphenyl)-, 3-(2'-methyl-4'-hydroxyphenyl)isocoumarins

etc. were prepared145 by condensation of homophthalic anhydride with

appropriate phenols. 3-(4'-Methoxyphenyl)isocoumarin was prepared by

condensation of homophthalic acid with anisole (Scheme 1.29).

COOH

COOH O

O

OO

/PPA

Scheme 1.29

1.6.5 Synthesis of isocoumarins via iodocyclization

A variety of 3-substituted 4-iodoisocoumarins and 6-substituted 5-iodo-

2(2H)-pyranones are readily prepared in excellent yields under mild reaction

conditions by the reaction of o-(1-alkynyl)benzoates and (Z)-2-alken-4-ynoates

with ICl146-148 (Scheme 1.30).

32

Page 52: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Scheme 1.30

OR1

R2

O

ICI O

O

IR2

OR1

R2

O

ICI O

O

IR2

1.6.6 Synthesis of isocoumarin by the use of new technology Microreactor technology has been studied as a suitable process to

produce chemicals via multicomponent reactions. Efforts were made to produce

3,4-diamino-1H-isochromen-1-ones. Based on a known reaction procedure,

using in situ generated HCN, a safe reaction setup was created to avoid the

release of the hazardous gas during the process. The 3,4-diamino-1H-

isochromen-1-ones149 were produced continuously in moderate to good yields

(Scheme 1.31).

NH

O

O

Scheme 1.31

OHH

O

O

OHH

N

O

R

RNH2KCN

HOAc O.

NH

O

R

N

HOAc

HNR

NH2

O

O

HNR

33

Page 53: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

1.7 Interconverision of isocoumarins and 3,4-dihydroisocumarins

We have seen so far 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.

1.7.1 Conversion of 3,4-Dihydroisocoumarins to Isocoumarins

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

dihydroisocoumarins to isocoumarins are as follows.

1.7.1.a Alkaline Hydrolysis Followed by Oxidation and Recyclization

Alkaline hydrolysis of 3,4-dihydroisocoumarins150 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 1.32).

O

O

RR

OH

RR

COOH

CrO3

O

RR

COOHO

O

RR

Scheme 1.32

1.7.1.b Benzylic Bromination Followed by Dehydrobromination

Isocoumarins can be prepared from 3,4-dihydroisocoumarin151-152 via

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

dehydrohalogenation with triethylamine (Scheme 1.33)

34

Page 54: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O

OO

O

O

OO

O

O

OO

Br

NBSUV

N(C2H5)3

Scheme 1.33

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.

1.7.2.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 hydroxy-acids. Cyclodehydration of the latter affords the

dihydroisocoumarins (Scheme 1.34)

O

O

Ar

OH

Ar

COOHO

Ar

COOH

O

O

Ar

O OO O

KOHCH3OH

NaBH4 -H20

Scheme 1.34

1.7.2.b Catalytic Reduction

Hydrogenation153-154 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.

35

Page 55: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

1.8 Reactions of isocoumarins and 3,4-dihydro- isocoumarin

1.8.1 Hydrolysis

Isocoumarins are lactones and undergo ring opening on alkaline

hydrolysis to give homophthaldehydic acids or ketones (106, R=H) or hydroxy

acid (107, R=alkyl, aryl, etc.). Similar treatment of 3,4-dihydroisocoumarins

yields the corresponding β-(2-carboxyphenyl)ethyl alcohol (107). 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 (108), the glycol produced recyclizes to the

more stable erythro-γ-lactone (109) under acid treatment155-156(Scheme 1.35).

OH

R'R

COOHO

R'R

COOH

O

O

OH

Ph

HH

COOOH

OHH

-

PhO

O

H H

OHPh

(106) (107)

(109)(108)Scheme 1.35

1.8.4 Reaction with Ammonia and Amines Ammonia and amines add to isocoumarins furnishing isocarbostyrils157

(110), a reaction typical of esters (Scheme 1.36).

O

O

NR

O

NH3orNH2R

(110)Scheme 1.36

36

Page 56: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

For example isocoumarin and 3-carboxylic acid have been condensed

with tryptamine, and the product subsequently converted to yobyrine (111) and

other derivatives158 (Scheme 1.37). .

Scheme 1.37

O

ON

NH2

N

ON

N

O

HN

+

(111)

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 (112) (Scheme 1.38).

.

O

OOH

HO

O

NH

OOH

HO

O

NH3

100°C

Scheme 1.38(112)

1.8.3 Reaction with Phosphorus Pentasulfide

Isocoumarin can be converted to 1-thioisocoumarin (113) with

phosphorus pentasulfide, and treatment of 1-thioisocoumarin with ammonium

sulfide or aniline yields isoquinolins (Scheme 1.39). Analogously, 3-

phenylisocoumarin has been converted to 1-thio-3-phenylisocoumarin, and

treatment with aniline produced (114)159.

37

Page 57: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O

O

S

P2S5

N N

O

Ph

Ph

(113) (114)

Scheme 1.39

1.8.4 Nitration

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

dihydroisocoumarin (115), in which nitric acid in sulfuric acid gives 3-(4-

nitrophenyl)-7-nitro-3,4-dihydroisocoumarin (116)160 (Scheme 1.40).

O

O

Ph

O

O

NO2

HNO3

O2N

(115) (116)Scheme 1.40

1.8.5 Reaction with Grignard Reagents

Addition of phenylmagnesium bromide161-166 to 3-phenylisocoumarin (117) followed by perchloric acid, anhydrous hydrochloric acid, ferric chloride or ferric

bromide yields the isobenzopyrilium salt (118) (Scheme 1.41).

O

O

Ph

O

Ph

O

Ph

Ph

PhHO

C6H5MgBr HY Y-

+

(117) (118)Scheme 1.41

Y= perchlorate, chloride, ferric chloride and ferric bromide, respectively.

38

Page 58: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

1.8.6 Oxidation

Chromium trioxide oxidation of 3,4,6,7-tetraphenylisocoumarin (119) produce 2-benzoyl-4,5-diphenylbenzoic acid161 (120) (Scheme 1.42).

O

Ph

Ph

PhPh

COOH

Ph

Ph

O

Ph

O

CrO3

(119) (120)Scheme 1.42

1.8.7 Reduction

The 3,4-double bond of isocoumarin is readily reduced with hydrogen and

palladium on charcoal or with other catalyst167-168. Catalytic reduction also has

been used to remove the halogen from cis- and trans-3-phenyl-4-halo-3,4-

dihydroisocoumarin169-170.

1.9 Plan of work More than thirty years elapsed, still increasing number of new

isocoumarins has been found in nature exhibiting a wide structural diversity in

their natural source and biosynthetic pathways. These findings are a constant

stimulant for synthetic work, which have been undertaken either to confirm novel

structures or to provide substantial amounts of material for biological and

pharmaceutical studies in those cases in which an isocoumarin exhibiting

interesting properties or was suspected of being responsible for the significant

properties associated with its natural source. In view of biological activity of

isocoumarin and 3,4-dihydroisocoumarin, it was planned to synthesize some of

outlined compounds. The work is divided in two groups.

i. Naturally occurring isocoumarin.

ii. Non naturally occurring isocoumarins and 3,4-dihydroisocoumarins.

39

Page 59: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

1.9.1 Naturally occurring isocoumarin In order to achieve the objective of the present work, a plan was made for

the synthesis of naturally occurring isocoumarin, Thunberginol B.

1.9.2 Unnaturally occurring isocoumarins and 3,4- dihydro isocoumarins

The plan for the preparation of unnatural isocoumarin and 3,4-

dihydroisocoumarin is to synthesize 3-dihalophenylisocoumarins and 3,4-

dihydroisocoumarins. Synthesis of all these compounds was based on the

following known principles, which are illustrated in (Scheme 1.43). i. Direct condensation of homophthalic acid with corresponding aryl

chlorides to obtain 3-substituted isocoumarins.

ii. Alkaline hydrolysis of the isocoumarin to obtain corresponding keto-acid.

iii. Reformation of the isocoumarin by refluxing of the keto-acid with acetic

anhydride.

iv. Preparation of recemic 3,4-dihydroisocoumarins by reduction of keto-

acids using sodium borohydride to yield corresponding hydroxy-acids

followed by cyclodehydration with acetic anhydride.

OHO

R

O

OHO

OH

O

200°C, 6h

OO

R

5% KOH/ C2H5OH

NaBH4

OHO

R

OH

OO

R

R Cl

O+

Ac2O

Ac2O

reflux

Scheme 1.43 General synthetic scheme for the synthesis of isocoumarins

and 3,4-dihydroisocoumarins

40

Page 60: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

42

Chapter-2 TOTAL SYNTHESIS OF THUNBERGINOL B

(A NATURAL ISOCOUMARIN)

Various naturally occurring isocoumarins have been isolated and synthesized

so far. In the present work, synthesis of thunberginol B has been carried out.

The detail of this work is as follows.

2.1 Synthesis of 6,8-dihydroxy-3-(3′,4′-dihydroxy-

phenyl)isocoumarin (Thunberginol B)

2.1.1 Introduction Thunberginols A (121), B (122), and F (125) were isolated from

Hydrangeae Dulcis Folium, the fermented leaves of Hydrangea macrophylla

SERINGE var. thunbergii MAKINO, as antiallergic constituents171-174.

Compounds 121, 122, and 125 inhibited histamine release from rat peritoneal

mast cells stimulated by compound 48/80, calcium ionophore A23187, or

antigen173-175 and oral administration of compound 121 inhibited passive

cutaneous anaphylaxis reactions in rats 176-177. However, their inhibitory

effects on the antigen-induced degranulation and release of cytokines in

basophils have not been reported to date.

Wang et al178 examined the effects of the isolated compounds

[thunberginols A (121), B (122), C (123), E (124), and F (125), phyllodulcin

(127), hydrangenol (128)] from Hydrangeae Dulicis Folium, thunberginol G

(126) and 3′-hydroxyhydrangeaic acid (129) derived from 127 on the

degranulations and/or releases of TNF-α and IL-4 via FcεRI signaling in rat

basophilic leukemia (RBL-2H3) cells. In addition, effects of the active

constituents on increase in [Ca2+]i were examined to get some information for

their mechanism of action. In conclusion, the 3-phenylisocoumarins

thunberginols A (121) and B (122)] and a benzylidene-phthalide thunberginol

F (125)] from the processed leaves of Hydrangea macrophylla var. thunbergii

(Hydrangeae Dulcis Folium) substantially inhibited the degranulation by

Page 61: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

43

antigen and calcium ionophore A23187, and the releases of TNF-α and IL-4

by antigen in RBL-2H3 cells. With regard to structural requirements of the 3-

phenylisocoumarins for the activity, the 3,4-double bond was essential for the

strong activity and the 6-hydroxyl group and lactone ring enhanced the

activity. The active compounds 121, 122 and 125 inhibited increase in [Ca2]i in

RBL-2H3 cells induced by antigen, but not by calcium ionophore A23187.

OOH

OH

OH O

OOH

OH

OH O

HO

O

OH

OH O

HO

OOH

O

OH O

HOH

OOH

OH

OH O

OOH

O

OH O

H

O

OH

OH O

OHOH

OH

OH O

OH

OH

OH

O

O

Thunberginol A (121) Thunberginol B (122) Thunberginol C (123)

Thunberginol G (126)Thunberginol F (125)Thunberginol E (124)

Phyllodulcin (127) hydrangenol (128) 3'-Hydroxyhydrangeaic acid (129)

Due to their biological importance, various routes were adopted to

synthesize thurnberginol B 179-180. We wish to report here a convenient route

for the synthesis of Thurnberginol B.

2.1.2 Plan of work

Keeping in mind the importance of Thunberginol B, It was planned to

synthesize it by the short and convenient route established M. Arfan et al. 3,5-

Dimethoxyhomophthalic acid is a key intermediate for the synthesis of highly

biological active naturally and unnaturally occurring isocoumarins and 3,4-

dihydroisocoumarins. It was synthesized efficiently in five steps from 3,5-

Page 62: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

44

dimethoxybenzaldehyde (130) via a series of reactions including synthesis of

3,5-dimethoxycinnamic acid181-182 (131) and 3-(3′,5′-dimethoxyphenyl)-

propionic acid183-184 (132). Cyclization of 3-(3′,5′-dimethoxyphenyl)propionic

acid (132) to 5,7-dimethoxy-1-indanone185-186 (133) and oxidative

decomposition of methyl 2-hydroxy-2-(5,7-dimethyoxy-1-oxo-1H-inden-2(3H)-

ylidene)acetate187(134) to 3,5-dimethoxyhomo-phthalic acid187-188 (135). 3,4-

Dimethoxybenzoylchloride (137) was prepared from 3,4-dimethoxybenzoic

acid (136) on reaction with thionyl chloride. Direct condensation of 3,4-

Dimethoxybenzoylchloride (137) with 3,5-dimethoxy- homophthalic acid (135) at 200oC afforded 3-(3',4'-dimethoxyphenyl)-6,8-dimethoxyisocoumarin (138). This isocoumarin (138) was purified by HPLC. Complete demethylation of

isocoumarin (138) with hydrobromic acid (48%) in acetic acid gave 3-(3',4'-

dihydroxyphenyl)-6,8-dihydroxyisocoumarin (139). Synthetic scheme is given

below (Scheme 2.1). 2.1.3 Results and discussion

Condensation of 3,5-dimethoxybenzaldehyde (130) with malonic acid

in the presence of pyridine (dry) and piperidine (dry) afforded 3,5-

dimethoxycinnamic acid (131), which showed a characteristic broad singlet at

δ 11.7 ppm for -OH in 1H NMR and C=C absorption at 1600 cm-1 in IR

spectrum. The detailed 1H NMR data is illustrated in (Table 2.1). The mass

spectrum of this compound showed molecular ion peak at m/z 208, which

agreed with its molecular weight. HREIMS of its molecular ion is in good

agreement with the calculated value. The fragmentation pattern of 3,5-

dimethoxycinnamic acid (131) is shown in scheme (Scheme 2.2). The

physical constants, IR and elemental analysis of all the synthesized

compounds is given in the experimental section.

Page 63: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

45

Na/Hg

MeONa / benzene

Diethyl oxalate

CH2(COOH)2

Pyridine(dry)/Piperidine(dry)

PPA

SOCl230min, 90°C

reflux6 hrs200°C

HBracetic acid

KOHH2O2

Thunberginol B

O

O

OO

O

COOH O

O

COOH

O

O O

O

O O

OH

COOCH3

O

O

COOH

COOH

OO

COOH

OO

COCl

O

O

O O

O

O

O

HO

OH O

OH

OH

(130) (131) (132)

(134)(133)

(136)

(135)

(137)

(138)(139)

[Yield: 86%][Yield: 83%]

[Yield: 77%][Yield: 91.5%]

[Yield: 72%]

[Yield: 60%]

[Yield: 53%]

Overall Yield = 0.86 × 0.83 × 0.77 × 0.915 × 0.72 × 0.60 × 0.53 ×100 = 11.5% Scheme. 2.1

This Part of the chapter has been published: Ghulam Qadeer. Nasim Hasan

Rama and Syed Jabbar Hussain Shah, “A new total synthesis of natural

isocoumarin, Thunberginol B”. ARKIVOC, 2007, (xiv), 12-19.

Page 64: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

46

O

O

COOHH

H

1'

2'1

2

34

56

(131) (132)

O

O

COOH1'

2'1

2

34

56

Table 2.1: 1H NMR Data of (131) & (132)

Carbon (131) (132) 2,6 6.66 (2H, dd, J = 1.9, 2.1 Hz) 6.36 (2H, dd, J = 2.1, 2.3 Hz)

4 6.5 (1H, t, J = 2.2 Hz) 6.32 (1H , t, J = 2.1 Hz)

1′ 7.69 (1H, d, J = 15.5 Hz) 2.89 (2H, t, J = 8.0 Hz)

2′ 6.4 (1H, d, J = 15.8 Hz) 2.66 (2H, t, J = 7.5 Hz)

OCH3 3.8 (6H, s) 3.76 (6H, s)

COOH 11.7 br s exchangeable with D2O 11.66 br s exchangeable with D2O

O

O

COOHO

O

OO COOH

O

O

O

O

O

O

O

-OH

-CO

-C2H2

-CH2O

-OCH3

-COOH

-C2H2

-CH2O

.

-OCH3

(m/z = 222, 8.58 % )

+

+

(m/z = 208, 100 % )

(m/z = 163, 15.17 % )

(m/z = 137, 6.98 % )

(m/z = 177, 9.87 % )

+

(m/z = 107, 3.78 % )(m/z = 77, 24.43 % )

(m/z = 106, 2.07 % )

(m/z = 132, 5.21 % )

(m/z = 191, 8.99 % )

+

+

-C2H2

(131)

++ .

..

.

-COOH.

+ .

.+

+

(m/z = 75, 3.81% )

+

Scheme 2.2: Mass Spectral Data and Fragmentation Pattern of (131)

Page 65: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

47

Reduction of cinnamic acid (131) with sodium amaglum yielded 3-

(3',5'-dimethoxyphenyl )propionic acid (132) which exhibited a broad singlet

at δ 11.66 ppm for COOH in 1H NMR which is exchangeable with D2O. The

carboxylic carbonyl absorption in IR spectrum was observed at 1722 cm-1.

The mass spectrum of the propionic acid (132) showed a molecular ion peak

at m/z 210 and a characteristic peak at m/z 165 [M+-COOH]. The

fragmentation pattern of propionic acid is illustrated in scheme (Scheme 2.3) and 1H NMR data is shown in (Table 2.1)

O

O

COOHO COOH-H2CO -COOH

+

+

(m/z = 210, 71 % )

(m/z = 150, 11.0 % )

(m/z = 105, 30.49 % )

(m/z = 165, 100 % )

(m/z = 139, 3.27 % )

(m/z = 180, 1.52 % )

O

O

O

O

-C2H4COOH-H2CO

+

+

(m/z = 65, 28.80 % )

(m/z = 91, 39.59 % )

+

(132)

+ .+ .

+ .

.

-CH2COOH.

-C2H2-COOH.

Scheme 2.3: Mass Spectral Data and Fragmentation Pattern of (132)

3-(3′,5′-Dimethoxyphenyl)propionic acid (132) on cyclization with poly-

phosphoric acid furnished the 5,7-dimethoxy-1-indanone (133) and its

structure was confirmed by the disappearance of broad singlet due to OH

proton (Exchangeable) and removal of one aromatic proton in 1H NMR while

carbonyl absorption in IR spectrum appeared at 1685 cm-1. Mass spectrum of

the indanone (133) showed molecular ion peak at m/z 192. The fragmentation

pattern of indanone is illustrated in scheme (Scheme 2.4) and 1H NMR data is

shown in (Table 2.2).

Page 66: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

48

O

O

O

OO

-OCH3

.

OO

O

O

O

O

O

O

O

OOO

++

+

+

+

+

++

(m/z = 134, 38.19 % )

(m/z = 106, 41.71% )

(m/z = 162, 36.01 % )

(m/z = 75, 16.68 % )(m/z = 103, 26.96 % )

(m/z = 131, 31.04 % )

(m/z = 163, 100 % )

(m/z = 105, 33.86 % )

(m/z = 192, 96.62 % ) (m/z = 150, 3.27 % )

(m/z = 119, 25.62 % )(m/z = 161, 90.11 % )(m/z = 133, 28.90 % )

-HCO.

-CH2CO

-CH2CO

-OCH3

.

-CO

-C2H4

-CO

-C2H4

-OCH3

.

-CO

-C2H4

+

(133)

. + .

+ . + .

+ .

-CH2O

Scheme 2.4: Mass Spectral Data and Fragmentation Pattern of (133)

5,7-Dimethoxy-1-indanone (133) on reaction with diethyl oxalate in

the presence of sodium methoxide in dry benzene gave methyl 2-hydroxy-2-

(5,7-dimethyoxy-1-oxo-1H-inden-2(3H)-ylidene)acetate (134) and its structure

was confirmed by 3H singlet for -COOMe at 3.95ppm in 1H NMR. IR spectrum

of this ester showed two additional bands at 3600-3000 cm-1 for OH and 1744

cm-1 for carbonyl of ester. Mass spectrum of (134) showed molecular ion peak

at m/z 278. The fragmentation pattern of indanone is illustrated in scheme

(Scheme 2.5) and 1H NMR data is shown in (Table 2.2).

Page 67: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

49

O

O

+

OO

OO

O

OH C2H3O2OO

O

OH

(m/z = 278, 18.90 % )

+

(134)

-CO

(m/z = 219, 96.33 % )

HO

(m/z = 191, 08.47 % )O

O

OO

O

OH

HO

O

O-

OO

O

(m/z = 191, 08.47 % )

O

O

(m/z = 278, 18.90 % )

-CO-CH2O

(m/z =133, 04.54% )

O

OH

HO

O

+ +

(m/z = 83, 100 % ) (m/z = 83, 100 % )

O

O HO

-CO

O

O

(m/z = 250, 02.10 % )

+

HOO

O

(m/z = 190, 03.17 % )

+ . .+ .

+ .

+ .

+ .

Scheme 2.5: Mass Spectral Data & Fragmentation Pattern of Compound (134)

Methyl-2-hydroxy-2-(5,7-dimethyoxy-1-oxo-1H-inden-2(3H)-ylidene)

acetate (134) on oxidation with hydrogen peroxide in the presence of

potassium hydroxide furnished 3,5-dimethoxyhomophthalic acid (135), which

showed a characteristic broad singlet at δ 11.2 ppm exchangeable with D2O

for -OH (Table 2.2). The two absorption bands of carbonyl groups appeared

at 1748 and 1697 cm-1 in IR spectrum. The mass spectrum of this compound

showed molecular ion peak at m/z 240, which agreed with its molecular

weight. The mass spectrum also showed two characteristic peaks at m/z =

Page 68: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

50

222, 195 [M+-H2O, -COOH] (Scheme 2.6). The structure of 3,5-

dimethoxyhomophthalic acid was also confirmed by X-ray crystallography.

1

2

34

56

7O

O

O

1

234

56

7O

O

O

OH

COOCH3

6

54

3

O

O

COOH

COOH

1'1

2

(133) (134) (135)

Table 2.2: 1H NMR Data of (133), (134) & (135)

Carbon (133) (134) (135)

2 2.9 (2H, t, J = 6.2 Hz) -- --

3 2.53 (2H, t, J = 6.1 Hz) 3.93 (2H, s) --

4 6.37 (1H, d, J = 1.4 Hz) 6.80 (1H, d, J = 3.0 Hz) 6.55 (1H , d, J = 1.2 Hz)

6 6.18 (1H, d, J = 1.6 Hz) 6.38 (1H,d, J = 3.05 Hz) 6.84 (1H , d, J = 1.4 Hz)

1′ -- -- 4.2 (2H, s)

OCH3 3.77 (3H, s)

3.80 (3H, s)

4.05 (6H, s)

3.65(3H, s )

3.67(3H, s)

COOCH3 -- 4.01 (3H, s) --

OH -- 13.57 (s) --

COOH -- -- 11.2 br. s exchangeable

with D2O

Page 69: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

51

OHO

OHO

O

O

-H2O

OHO

O

O-CO2 O

O

O

O

O

O

OO

OOHO

O

O

-CO

O

O

O

OO

-CO

(m/z = 194, 57.79 % )(m/z = 195 , 12.77 % )

m/z = 196 ( 56.89 % ) (m/z = 222, 8.58 % )

(m/z = 151, 14.85 % )(m/z = 178, 100 % )

(m/z = 150, 2.51 % )

(m/z = 120, 3.55 % )

CO O

O

.

-COOH.

.

+

+

+

-CO2-COOH.

-OCH3

(m/z = 240, 21.69 % )(135)

+ . + .

+ .

+ .

+ .

+ .

Scheme 2.6: Mass Fragmentation Pattern of Compound (135)

The X-ray crystal structure of (135) is shown in Fig. 2.1. The C9/O3/O4

carboxyl group is tilted by 30.34 (9) with respect to the plane of the benzene

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama and Fan Zhijin, “Synthesis, mass fragmentation studies and bioloigical

evaluation of 3,5-dimethoxyhomophthalic acid and related compounds”. Chem.

Res. Chinese U., 2007, 23(5), 533-537.

Page 70: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

52

ring, possibly as a result of the steric hindrance of the ortho substituents. The

O3—C9—C2—C3 and O3—C9—C2—C1 torsion angles are 149.68 (14) and

29.6 (2), respectively.

Figure 2.1: View of the 3,5-dimethoxyhomophthalic acid, with displacement

ellipsoids drawn at the 30% probability level

.

It is noteworthy that the C9 and O3 carbonyl group derivatives from the

phenyl plane, with the mean deviation from the plane of 0.1750\%A. The

torsion angles of O3-C9-C2-C3 (149.68(14)\%) and O3-C9-C2-C1 (-

21.6(2)\%) have also indicated this carbonyl group π system is not coplanar

with the phenyl plane. These results display that some repulsions among

substitutes may exist. It is also noted that this compound forms an infinite 2D

network structure by the intermolecular hydrogen bonds between C7-

H7A...O3 (2.7829 (19)\%A), C10-H10C...O2 (3.4144 (19)\%A), O4-H4A...O3

(2.6606 (14)\%A) and O1-H1...O2 (2.6532 (14)\ %A) (Fig. 2.2). In the crystal

packing, intermolecular C—H….O hydrogen-bond interactions link the

molecules, forming a two-dimensional network (Fig. 2.2).

Page 71: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

53

Figure 2.2: The crystal packing of the 3,5-dimethoxyhomophthalic acid,

viewed approximately along a axis. Hydrogen bonds are shown as dashed

lines

[symmetry codes: (a)1x, 1y, 2z (b) x, 2y, 2z (c) x, 1y, 1z]

Crystal data

Mr = 240.21 Triclinic, P1 a = 7.2886 (10) Å b = 7.9631 (11) Å c = 10.2758 (14) Å α = 105.937 (2)º β = 103.444 (2)º γ = 93.211 (2)º

V = 553.17 (13) Å3 Z = 2 Mo Kα µ = 0.12 mm−1 T = 292 (2) K 0.24 × 0.20 × 0.20 mm

This Part of the chapter has been published: Ghulam Qadeer. Nasim Hasan

Rama and Qing-Shan Li. “Crystal structure of 2-(Carboxymethyl)-4,6-

dimethoxybenzoic acid”. Acta Cryst. 2006, E62, 906–907.

Page 72: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Geometric parameters (°A, °)

Selected bond lengths Selected bond angles

C1—C6 1.3743 (19) C1—C2 1.4128 (18) C1—C7 1.5131 (18) C9—O3 1.2346 (17) C2—C3 1.4085 (18) C9—O4 1.2979 (17) C2—C9 1.4822 (18) C11—O6 1.4243 (18) C5—O6 1.3625 (16) C5—C6 1.3888 (19

Hydrogen-bond geometry (Å, °)

D—H···A D—H H···A C7—H7B···O3 0.97 2.43 C10—H10C···O2i 0.96 2.56 O4—H4A···O3ii 0.850 (10) 1.821 (O1—H1···O2iii 0.853 (10) 1.801 (

Symmetry codes: (i) −x, −y+1,−y+1, −

3,4-Dimethoxybenzoylchloride (dimethoxybenzoic acid (136) on rea

condensation of 3,4-dimethoxybenz

dimethoxyhomophthalic acid (135) at 2

dimethoxyphenyl)isocoumarin (138), absoption at 1716 and C=C absorbtion

characteristic C4-H singlet at δ 7.0 ppm

data is illustrated in (Table 2.3). The

showed molecular ion peak at m/z 34

weight. The mass spectrum also showe

254, 164 (Scheme 2.7). This isocoumari

53

C6—C1—C2 120.35 (12) C6—C1—C7 116.69 (12) O2—C8—O1 122.97 (13) C2—C1—C7 122.77 (12) O2—C8—C7 124.33 (12) C1—C2—C9 119.88 (12) C4—C5—C6 120.42 (12) C8—C7—C1 114.89 (11) C8—C7—H7A 108.5 C8—O1—H1 108.8 (16)

D···A D—H···A

2.7829 (19) 101 3.4144 (19) 148 11) 2.6606 (14) 169 (2) 10) 2.6532 (14) 178 (2)

−z+1; (ii) −x, −y+2, −z+2; (iii) −x+1, z+2

137) was prepared from 3,4-

ction with thionyl chloride. Direct

oylchloride (137) with 3,5-

00oC afforded 6,8-dimethoxy-3-(3',4'-

which showed lactonic carbonyl

at 1599 cm-1 in IR spectrum and

in 1H NMR. The detailed 1H NMR

mass spectrum of this compound

2, which agreed with its molecular

d two characteristic peaks at m/z =

n (138) was purified by HPLC.

Page 73: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O

O O

O

O

12

345

67

8

1' 2'

3'

4'5'

6'

(138) (139)

O

HO

OH O

OH

OH

12

345

67

8

1' 2'

3'

4'5'

6'

Table 2.3: 1H NMR and 13 C NMR data of (138) and (139)

(138)

(139)

Carbon 1H NMR 13 C NMR 1H NMR 13 C NMR

1 --- 154.9 --- 151.9 3 --- 149.2 --- 147.2 4 7.0(1H,s) 111.3 6.91(1H,s) 111.3 4a --- 148.7 --- 148.7

5 7.55 (1H, d, J = 1.8 Hz) 112.2 7.47 (1H, d, J = 1.8

Hz) 115.2

6 --- 167.6 167.6

7 7.44(1H, d, J = 1.8 Hz) 111.8 7.41(1H, d, J = 1.8

Hz) 113.8

8 --- 162.8 --- 162.8 8a --- 112.6 --- 112.6 1′ --- 125.4 --- 125.4

2′ 7.57(1H, d, J = 1.8 Hz) 120.5 7.52(1H, d, J = 1.8

Hz) 122.5

3′ --- 154.9 --- 154.9 4′ --- 154.9 --- 154.9

5′ 7.16(1H, d, J = 8.7 Hz) 123.4 7.11(1H, d, J = 8.7

Hz) 125.4

6′ 7.76(1H, dd, J = 8.4, 1.2Hz) 123.6 7.72 (1H, dd, J =

8.4,1.2 Hz) 124.6

OCH3 3.79-3.88 (12H, 4×s) 55.8-56.3 --- --- OH --- --- 5.22(s) ---

54

Page 74: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O

O

O

O

OO

O

O

O

O

O

O

O

O

O

OO

O

O

-CO

-CO

-CO2-OCH3

O-

.

-HCO

(m/z = 138, 21 %)

(m/z = 166, 7.0 %)(m/z = 342, 100 %)

(m/z = 300, 2.2 %)

(m/z = 284, 30 %)

(m/z = 314, 30 %)

(m/z = 178, 01 %)(m/z = 206, 32 %)

OO

O

O

O

O

O

O

O

O

O

-.O

O

(138)

+

+ .++ .

+ .

++

.+

Scheme 2.7: Mass Fragmentation Pattern of Compound (138)

Complete demethylation of this 6,8-dimethoxy-3-(3',4'-dimethoxy-

phenyl)isocoumarin (138) with hydrobromic acid (48%) in acetic acid gave 3-

(3′,4′-dihydroxyphenyl)-6,8-dihydroxyisocoumarin (139). Its IR spectrum

showed a broad absoption at 3386-3360cm-1 for phenolic OH group. In 1H

NMR, the signal for methoxy groups in (138) at δ 3.83 ppm disappeared. The

detailed 1H NMR data is illustrated in (Table 2.3). The mass spectrum of this

compound showed molecular ion peak at m/z 286, which agreed with its

molecular weight. The mass spectrum also showed two characteristic peaks

at m/z = 227, 136 (Scheme 2.8). This isocoumarin (139) is also purified by

HPLC.

55

Page 75: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O

OH

HO

OH

OH

OH

OH

OH

HO

OH

OH

OOH

OH

OH

OHO

OH

OH

O

O

HO

OH

O

OH

HO

HO

OH

OOH

HO

OH

-CO

-CO

-CO2-OH

-.

O-

.

-HCO

.

-.OH

OH

(m/z = 110, 32 %)

(m/z = 138, 7.0 %)(m/z = 286, 7.0 %)

(m/z = 258, 2.2 %)

(m/z = 136, 100 %)

(m/z = 243, 27 %)

(m/z = 259, 52 %)

(m/z = 150, 01 %)(m/z = 178, 31 %)

+

+

(139)

+

+ .

+

+ .

.+ .

+

+

Scheme 2.8: Mass Fragmentation Pattern of Compound (139)

2.1.4 Experimental

All reagents and solvents were commercially available and used

as supplied. The melting points of the compounds were determined in open

capillaries using a Gallenkemp melting point apparatus and are uncorrected.

The infrared spectra were recorded on a Hitachi model 270-50

spectrophotometer as KBr disks or as neat liquids. 1H NMR (300 MHz) and 13C NMR (75 MHz) spectra were recorded on a Bruker AM-300 in CDCl3

56

Page 76: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

solution using TMS as an internal standard. HPLC-MS analyses were

performed HPLC-Anlage Waters 2695 Alliance, Säule Phenomenex Luna 3µ

Phenyl-Hexyl (2 × 150 mm), mobile Phase Water/Acetonitrile (0:100 bis

50:50), Fluss: 0.2 ml/min, photodiode array detector Waters 996, MS-Detector

Waters ZQ2000. In the mass detector, the fragmenter operated at 70eV.

Elemental analysis was performed on a LECO CHNS-932 instrument. XRD

data collection was carried out by, Data collection: SMART (Bruker, 1998);

cell refinement: SAINT (Bruker, 1999); data reduction: SAINT; program(s)

used to solve structure: SHELXS97 (Sheldrick, 1997); program(s) used to

refine structure: SHELXL97 (Sheldrick, 1997); molecular graphics: SHELXTL

(Bruker, 1999); software used to prepare material; SHELXTL.

3, 5-Dimethoxycinnamic acid (131)

3,5-Dimethoxybenzaldehyde (130) (17g, 102 mmol), malonic acid

(51g), dry pyridine (183.6mL) and dry piperidine (40.2mL) were heated on

boiling water bath for 4h. The reaction mixture was cooled and poured into

cold water (500mL) and carefully acidified with dilute hydrochloric acid (1:1)

with stirring. On filtration white ppt of 3,5-dimethoxycinnamic acid (131)(18.3g,

88 mmol) was obtained. Yield: 86%; m.p: 170°C; IR (KBr, νmax, cm-1): 3354,

1685,1600; 1H NMR (CDCl3, δ ppm): 7.69 (1H, d, J =15.5 Hz, H-1′), 6.66 (2H,

d, J = 2.1 Hz, H-2,6), 6.50 (1H, t, J = 2.2 Hz, H-4), 6.41 (1H, d, J = 15.8 Hz, H-

2′), 3.84 (6H, s, OCH3), 11.70 (bs COOH); EIMS (DMF, m/z, %): 208 (100,

M), 191 (9), 177 (10), 163 (15), 137 (7), 132 (5), 107 (4), 106 (2), 77 (24), 75

(4), 51 (24); Anal. Cald for C11H12O4 : C, 63.45; H, 5.81; Found: C, 63.01; H,

5.54.

3-(3′,5′-Dimethoxyphenyl)propionic acid (132)

Cinnamic acid (131) (17.85g, 86 mmol) was dissolved in 93mL of IM

solution of sodium hydroxide contained in a 500ml two necked flask equipped

with a mechanical stirrer and added sodium amalgam (222g) gradually during

1 hour while the reaction mixture was well stirred. When hydrogen was not

57

Page 77: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

evolved, the mercury was separated and washed with water, washings was

added to the solution and acidified the reaction mixture with dilute

hydrochloric acid (1:1). Propionic acid was precipitated at first in the form of

an oil, which solidified on cooling and rubbing with a glass rod. Filtered at the

pump and recrystalized with pet. ether to get 3-(3′,5′-Dimethoxyphenyl)-

propionic acid (132) (15g, 71mmol). Yield: 83%; m.p: 58°C; IR (KBr, νmax, cm-

1): 3390, 1722, 1207; 1H NMR (CDCl3, δ ppm): 6.36 (2H, d, J = 2.3 Hz, H-2,6),

6.32 (1H, t, J = 2.1 Hz, H-4), 2.89 (2H, t, J = 8.0 Hz, H-1′), 2.66 (2H, t, J = 7.5

Hz, H-2′), 3.76(6H, s, OCH3), 11.66 (bs COOH); EIMS (DMF, m/z, %): 210

(71, M), 180 (2), 165 (100), 150 (11), 139 (3), 105 (30), 91 (40), 65 (29); Anal.

Cald for C11H14O4 : C, 62.85; H, 6.71; Found: C, 62.39; H, 6.14.

5,7-Dimethoxy-1-indanone (133) 3-(3,5-Dimethoxyphenyl)propionic acid (132) was dissolved in 250g of hot

poly phosphoric acid (90°C). The resulting yellow solution was heated on oil

bath with stirring for 2 hours. The cooled solution was added to 500ml of ice

water. Extracted the mixture with three 150ml portions of ethyl acetate and

washed the combined extracts with 5% sodium hydroxide solution and then

with water until the washings were neutral. Dried the ethyl acetate solution

over anhydrous magnesium sulphate. The organic layer was concentrated

and chromatographed on silica gel using pet. ether (40-80°C) as eluent to

afford 5,7-dimethoxy-1-indanone (133) (8.8g, 6 mmol). Yield: 77%; m.p: 97-

98°C; IR (KBr, νmax, cm-1): 1685, 1209,1057; 1H NMR (CDCl3, δ ppm): 6.18

(1H, d, J = 1.6 Hz, H-6), 6.37 (1H, d, J =1.4 Hz, H-4), 2.9 (2H, t, J = 6.2 Hz, H-

2), 2.53 (2H, t, J = 6.1 Hz, H-3), 3.80 (3H, s, OCH3), 3.77 (3H, s, OCH3);

EIMS (DMF, m/z, %): 192 (97, M), 163 (100), 162 (36), 161 (90), 150 (3) 134

(38), 133 (29), 131(31), 119 (25), 105 (34), 106 (42), 103 (30), 75 (17); Anal.

Cald for C11H12O3: C, 68.74; H, 6.29; Found: C, 68.11; H, 6.94.

58

Page 78: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Methyl 2-hydroxy-2-(5′,7′-dimethyoxy-1-oxo-1H-inden-2(3H)-ylidene)-

acetate (134)

Sodium (15.2g) was dissolved in methanol (27.5g) and benzene (440

ml) was added to this solution. Indanone (133) (20g, 100 mmol) was added in

portions with good stirring, then diethyl oxalate (38.5mL) was added as single

aliquot and the reaction mixture was stirred for over night at room

temperature. Hydrochloric acid (5%) was added under ice cooling to the

residue obtained after removal of the solvent, to give a final pH of 1.0. The

precipitate that deposited was collected on a filter, washed with hydrochloric

acid (5%), then with water, and dried. The solid thus obtained was crystallized

from methanol to give methyl 2-hydroxy-2-(5,7-dimethyoxy-1-oxo-1H-inden-

2(3H)-ylidene)acetate (134) (26.5g, 90 mmol) as yellow needles. Yield:

91.5%; m.p: 234-237 °C; IR (KBr, νmax, cm-1): 3400-2400, 1744 ,1692, 1633,

1605, 1365; 1H NMR (CDCl3, δ ppm): 13.57 (s, OH), 6.80 (1H, d, J = 3.0 Hz,

Ar-H), 6.38 (1H,d, J = 3.05 Hz, Ar-H), 4.05 (6H, s, 2 × OMe), 4.01 (3H, s,

COOMe), 3.93 (2H, s, CH2); EIMS (DMF, m/z, %): 278 (19, M), 219(95),

191(8), 175(6), 85(65), 83(100), 42 (25). Anal. Cald for C14H14O6: C, 60.43; H,

5.07; Found: C, 60.08; H, 5.43.

3,5-Dimethoxyhomophthalic acid (135)

A solution of methyl 2-hydroxy-2-(5,7-dimethyoxy-1-oxo-1H-inden-

2(3H)-ylidene)acetate (134) (26.5g, 100 mmol) and potassium hydroxide

(20.8g) in water (442mL) was cooled to 5-10°C, and hydrogen peroxide

(90.0g, 35%) was added. The reaction mixture was stirred at this temperature

for 6 hours. When the temperature was gradually raised to 50°C, vigorous

frothing occurred. After stirring for further 2h at this temperature, the reaction

mixture was filtered and the filtrate was acidified with concentrated

hydrochloric acid. The crystals that precipitated were collected by filtration

and the filtrate was extracted with ethyl acetate (4 × 30mL). The solvent was

evaporated to give solid residue, which was purified by recrystalization from

dichloromethane to give 3,5-dimethoxyhomophthalic acid (135) (19g, 72

59

Page 79: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

mmol) as light yellow color micro crystals. Yield: 72%; m.p: 170°C; IR (KBr,

νmax, cm-1): 2580-3300, 1714, 1659, 1226; 1H NMR (CDCl3, δ ppm): 11.7 (bs

COOH), 6.84 (1H, d, J = 1.4 Hz, Ar-H), 6.55 (1H, d, J = 1.2 Hz, Ar-H), 4.2 (2H,

s, H-1′), 3.67 (3H, s, OCH3), 3.65 (3H, s, OCH3); EIMS (DMF, m/z, %): 240

(38, M), 222 (17), 196 (66), 195 (40), 194 (85), 178 (100), 151 (28), 150 (15),

120 (41), 119(12). Anal. Cald for C11H12O6: C, 55.00; H, 5.04; Found: C,

54.52; H, 5.41.

3-(3′,4′-Dimethoxyphenyl)-6,8-dimethoxyisocoumarin (138)

Mixture of 3,4-dimethoxybenzoic acid (135) (2g, 10.9 mmol) and thionyl

chloride (1.57g, 1mL, 13.2 mmol) was heated for 30 min in the presence of a

drop of dimethylformamide at 90°C. Completion of the reaction was indicated

by the stoppage of gas evolution. Removal of excess of thionyl chloride was

carried out under reduced pressure to afford 3,4-dimethoxybenzoyl chloride

(137) (1.8g, 9mmol). 3,5-Dimethoxyhomophthalic acid (136) (0.63g, 2.6

mmol) and 3,4-dimethoxybenzoyl chloride (137) (1.8g, 9 mmol) were refluxed

at 200°C for six hours. Residue, after concentration, was purified by HPLC to

give the 3-(3′,4′-dimethoxyphenyl)-6,8-dimethoxyisocoumarin (138). Yield:

60%; m.p: 175-177°C; IR (KBr, νmax, cm-1): 1716 (C=O, lactonic), 1599 (C=C); 1H NMR (CDCl3, δ ppm): 7.76(1H, dd, J = 1.2, 8.4 Hz, H-6′), 7.57(1H, d, J =

1.8 Hz, H-2′), 7.55 (1H, d, J= 1.8 Hz, H-5′), 7.44 (1H, d, J= 1.8 Hz, H-7),

7.16(1H, d, J= 8.7 Hz, H-5′),7.0(1H, s, H-4), 3.88(3H, s), 3.85(3H, s),

3.82(3H, s), 3.79 (3H, s); 13C NMR (CDCl3, δ ppm): (167.6, 162.8, 154.9,

149.2, 148.7, 125.4 , 123.6, 123.4, 112.6, 112.2, 111.8)(Ar-C), 55.8-56.3 (4×

OCH3); EIMS (DMF, m/z, %): 342(6.0, M), 314(30), 300(2), 284(30), 254(54),

206(32), 178(1), 166(7), 164(100), 138(21), 94(13); Anal. Cald for C19H18O6:

C, 66.66; H, 5.30; Found: C, 66.20; H, 5.05.

60

Page 80: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

3-(3′,4′-Dihydroxyphenyl)-6,8-dihydroxyisocoumarin (139)

Freshly distilled hydrobromic acid (55%, 16 mL) was added to stirred

solution of isocoumarin (138) (0.3g, 1.1 mmol) in glacial acetic acid (16mL).

Reaction mixture was then refluxed for four hours, cooled and poured on

crushed ice and then treated with solid sodium carbonate till PH-7 and

extracted with diethyl ether (2 × 30 mL), dried over anhydrous sodium

sulphate and concentrated to give crude solid which was purified by HPLC to

get 3-(3′,4′-dihydroxyphenyl)-6,8-dihydroxyisocoumarin (139) Yield: 53%;

m.p: 247–249°C(lit. mp 244°C); IR (KBr, νmax, cm-1): 1716 (C=O, lactonic),

1599 (C=C); 1H NMR (CDCl3, δ ppm): 7.69(1H, dd, J = 1.2, 8.4 Hz, H-6′),

7.52(1H, d, J = 1.8 Hz, H-2′), 7.49 (1H, d, J = 1.8 Hz, H-5′), 7.38(1H, d, J =

1.8 Hz, H-7), 7.19(1H, d, J = 8.7 Hz, H-5′), 6.94(1H, s, H-4); 13C NMR

(CDCl3, δ ppm): 166.9, 164.8, 157.1, 148.2, 147.7, 128.4, 124.6, 122.4, 119.5,

111.6, 110.2, 109.8; EIMS (DMF, m/z, %): 286 (52), 269 (100), 259 (4), 241

(13), 213 (11), 177 (23), 149 (3), 137 (4), 121 (7); Anal. Cald for C15H10O6: C,

62.94; H, 3.52; Found: C, 62.71; H, 3.84.

61

Page 81: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Chapter-3 SYNTHESIS OF UNNATURAL ISOCOUMARINS

3.1 Synthesis of dihalophenylisocoumarins

3.1.1 Introduction Naturally occurring isocoumarins containing halogens have been

seldom reported. Examples of naturally occurring isocoumarins containing

fluorine are not known yet. However, a few examples of naturally occurring

chlorine- and bromine-containing isocoumarins have been reported.

Laresenb189 has isolate chlorine-containing metabolite dichlorodiaportin (140) from the cheese-associated cultures of Penicillium nalgiovense. 4-Chloro-3-

(4′-fluorobenzyloxy)isocoumarin (141) has been found190 to be quite an

effective inhibitor for human Q31 granzyme A, murine and human granzyme

A, isolated from cytotoxic T lymphocytes. This isocoumarin derivative has also

been found191, to be useful in the treatment of emphysema as serine protease

inhibitor.6-(2′-Chloro-4′-trifluoromethylphenoxy)-3,4-dihydroisocoumarin (142)

has been used192, as a herbicide, which almost totally controlled the growth of

Schinochloa crusgall, Sinapis alba and other weeds. 7-Amino-4-chloro-3-(2′-

bromoethoxy)isocoumarin (143) has been synthesized,193 and evaluated as a

potent inhibitor of human leuko elastase and several blood coagulation

enzymes194. 7-Amino-3-(2′-bromopropoxy)-4-chloroisocoumarin and 7-Amino-

3-(3′-bromopropoxy)-4-chloroisocoumarin have been patented195, as an

ascapain inhibitor in the inhibition and treatment of neurodegeneration.

O

O

ClO

F

O

O

OCl

F3CO

O

OBr

Cl

H2N

O

Cl

ClO

OH O

OH

(140)(141)

(142)(143)

62

Page 82: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

3.1.2 Plan of work

As a continuation of our previous studies196-197 and biological activities

associated with halo substituted isocoumarins, prompted us to synthesize

some new 3-(dihalophenyl)isocoumarins 146(a-g) and their conversion to the

corresponding 3,4-dihydro-derivatives 149(a-g) in order to check their

antimicrobial, anti-inflammatory, antioxidant, antiviral, anti-HIV, anti-HBV,

pesticide and plant disease control activities. Synthetic scheme is shown as

follows (Scheme 3.1)

OHO

Ar

O

OHO

OH

O

Ar OH

O

SOCl2Ar Cl

O

200°C, 6h

OO

Ar

5% KOH/ C2H5OH

NaBH4 /H+

OHO

Ar

OH

OO

Ar

FF

F

F

F

F

b ) Ar =a) Ar = d) Ar =c) Ar =F

F

Cl

Cl ClCl

Cl

Cl

e) Ar = f) Ar = g) Ar =

Ar Cl

O+

Ac2O

Ac2O

30 minreflux

reflux

148(a-g) 147(a-g)149(a-g)

146(a-g)

144(a-g) 145(a-g)

145(a-g)[Yield = 77- 81%]

[Yield = 69 - 81%]

[Yield = 69 - 79%][Yield = 69- 80%]

Scheme 3.1

3.1.3 Results and Discussion

Difluorobenzoic acids 144(a-d) and dichlorobenzoic acids 144(e-g) were converted into their respective acid chlorides 145(a-g) by reaction with

thionyl chloride in the presence of a drop of DMF. Direct condensation of acid

63

Page 83: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

chlorides 145(a-g) with unsubtituted homophthalic acid at 200oC afforded 3-

(dichlorophenyl)isocoumarins 146(a-d) and 3-(difluorophenyl)isocoumarins 146(e-g) These isocoumarins 146(a-g) were purified by column

chromatography and showed a single spot on TLC.These isocoumarins

146(a-g) exhibited characteristic 1H singlet at δ 6.81-7.19 ppm for C4-H in 1H

NMR (Table 3.1). The aromatic protons appeared in the acceptable region i.e.

7.50-8.31 ppm. In IR spectra of isocoumarins 146(a-g) lactonic carbonyl

absorptions were observed at 1702-1709 cm-1. The molecular ion peaks in

the mass spectrum of 3-(3′,5′-difluorophenyl)isocoumarin 146a and for 3-

(3′,5′-dichlorophenyl)-isocoumarin 146e were obtained at m/z 258 and 292

respectively (Scheme 3.2 & 3.3)

O

O1

2

3456

78

1' 2'

3'4'5'

6'

146a

F

F

Table 3.1: 1H NMR Data of 3-(3′.5′-Difluorophenyl)isocoumarin (146a)

1H NMR δ (ppm) Carbon

No 3-(3′.5′-Difluorophenyl)isocoumarin

146a

4 6.94 (s)

5 7.54 (d, J = 7.40 Hz)

6 7.74 (1H, m)

7 7.50 (dd, J = 7.88 Hz)

8 8.31 ( d, J = 7.92 Hz

2′ 7.40 ( dd, J = 2.0, 8.30 Hz)

3′ ---

4′ 6.81 ( m)

5′ ---

6′ 7.40 ( dd, J = 2.0, 8.30 Hz)

64

Page 84: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OO

O

-CO

-CO

-F-CO2

-.

O-

.

+

+

+

OO F

F

F

F

F

F

O

F

F

OF

FF

.

O

F

F

F

F

-HCO

(m/z = 258, 56.72 %) (m/z = 230, 30.88 %)(m/z = 141, 7.74 %)

(m/z = 113, 32.74 %)(m/z = 216, 2.7 %) (m/z = 212, 24.83 %)

(m/z = 139, 100%)

(m/z = 117, 2.61 %)(m/z = 145, 34.75 %)

+

(146a)

+ .+ .

+

+ + ..

Scheme 3.2: Mass Fragmentation Pattern of 3-(3′,5′ Difluorophenyl)-

isocoumarin 146a

65

Page 85: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O1

2

3456

78

1' 2'

3'4'5'

6'

146e

Cl

Cl

Table 3.2: 1H NMR Data of 3-(3′.5′-Dichlorophenyl)isocoumarin (146e)

1H NMR δ (ppm)

Carbon No

3-(3′.5′-Dichlorophenyl)isocoumarin

146e

4 6.95 (s)

5 7.55 (1H, dd, J 2.2, 7.6 Hz)

6 7.53 (m)

7 7.50 (m)

8 8.31 (dd, J = 1.9, 7.9 Hz)

2′ 7.75 (t, J = 1.2 Hz)

3′ ----

4′ 7.39 (t, J = 1.2 Hz)

5′ ----

6′ 7.75 (t, J = 1.2 Hz)

66

Page 86: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OO

O

-CO

-CO

-Cl-CO2

-.

O-

.O

O Cl

Cl

Cl

Cl

Cl

Cl

O

Cl

Cl

O

Cl

ClCl

.

O

Cl

Cl

Cl

Cl

-HCO

m/z = 290 (56.72 %, 2 Cl35) 292 (38.18 %, Cl35Cl37) 294 (6.52 %, 2 Cl37)

m/z = 262 (30.88 %, 2 Cl35) 264(20.28 %, Cl35Cl37) 266 (3.59 %, 2 Cl37)

m/z = 174 (7.74 %, 2 Cl35) 176 (4.84 %, Cl35Cl37) 178 (0.75 %, 2 Cl37)

m/z = 145 (32.74 %, 2 Cl35) 147 (20.85 %, Cl35Cl37) 149 (3.85 %, 2 Cl37)

m/z = 246 (2.7 %, 2 Cl35) 248 (1.6 % Cl35Cl37) 250 (0.3 % 2 Cl37)

m/z = 227 (24.83 %, 2 Cl35) 229 (8.23 %, Cl35Cl37)

m/z = 173(100%, 2 Cl35) 175(64.21 %, Cl35Cl37) 177(10.32 %, 2 Cl37)

m/z = 117 (0.61 %)m/z = 145 (32.74 %)

(146e)

++

+

+ .+ .

+

+ .

+

+

.

Scheme 3.3: Mass Fragmentation Pattern of 3-(3′,5′-Dichlorophenyl)-

isocoumarin 146e

Alkaline hydrolysis of isocoumarins 146(a-g) afforded 2-(difluoro-

benzoylmethyl)benzoic acids 147(a-d) and 2-(dichlorobenzoylmethyl)-

benzoic acids 147(e-g). In the IR spectra of keto-acids 147(a-g), the

absorptions for the ketonic carbonyl were observed at 1708-1713cm-1.The

keto-acids 147(a-g) showed 2H singlets at δ 4.60-4.65 ppm for methylene

protons at C-1′ (Table 3.3 & 3.4). The molecular ion peak for the keto-acids

147(a-d) and for 147(e-g) were observed at m/z 276 (M+) and 308(M+)

67

Page 87: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

(Scheme 3.4 & 3.5). Isocoumarins 146(a-g) can also be obtained on

refluxing keto-acids 147(a-g) with acetic anhydride (reversible reaction).

COOHO

2'1'34

56

1'' 2''3''

4''5''6''

1

2

147a

F

F

Table 3.3: 1H NMR Data of 2′-(3′′,5′′-Difluorobenzoylmethyl)benzoic

acid (147a)

1H NMR δ (ppm) Carbon

No 2′-(3′′,5′′-difluorobenzoylmethyl)benzoic acid

147a

3 7.23 (d, J = 7.4Hz)

4 7.37 (dd, J = 7.6 Hz)

5 7.77 (m)

6 8.08 (d, J = 7.23 Hz)

1′ 4.61 (s)

2′′ 7.51 (m)

3′′ ----

4′′ 6.98 ( dd, J = 8.25, 16.6 Hz)

5′′ ----

6′′ 7.51 (m)

COOH 11.31 (s) Exchangeable with D2O

68

Page 88: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OO

F

-H2O

O OH

OF

O OH

O

OF

F

F

OO

OF

O

2(-F)

-CO

-C2H2

-CO2

F

-

F.-CO.-

-C2H2

F

(m/z = 290, 22 %)

(m/z = 164, 4.20 %)

(m/z = 145, 63.99 %)

(m/z = 101, 4.22 %)

(m/z = 75, 22.71 %)(m/z = 224, 22 %)

(m/z = 276, 2.5 %)

(m/z = 158, 17.39 %)

(m/z = 262, 1 %)

(m/z = 109, 100 %)(m/z = 113, 63.99%)F

F

F

F

F

F

(147a)

+ + +

+ .+ .

+ .

+ . + .+ .

+.

+ .

Scheme 3.4: Mass Fragmentation Pattern of 2′-(3′′,5′′-

Difluorobenzoylmethyl) benzoic acid 147a

69

Page 89: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

COOHO

2'1'3

4

56

1'' 2''

3''

4''5''6''

1

2

147e

Cl

Cl

Table 3.4: 1H NMR Data of 2′-(3′′,5′′-Dichlorobenzoylmethyl)benzoic

acid (147e)

1H NMR δ (ppm) Carbon

No 2′-(3′′,5′′-dichlorobenzoylmethyl)benzoic acids

147e

3 7.18 (dd, J = 1.56, 7.99 Hz )

4 7.24 (m)

5 7.52 (m)

6 8.15 (dd, J = 1.2, 7.9 Hz

1′ 4.62 (s)

2′′ 7.94 (d, J = 1.76 Hz

3′′ ----

4′′ 7.56 (dd, J= 2.41 Hz

5′′ ----

6′′ 7.94 (d, J = 1.76 Hz

COOH 10.31 (s) Exchangeable with D2O

70

Page 90: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OO

Cl

-H2O

O OH

OCl

O OH

O

OCl

Cl

Cl

OO

OCl

O

2(-Cl)

-CO

-C2H2

-CO2

Cl

-

Cl.

-CO.-

-C2H2

++

+

Cl

(m/z = 290, 22 %, 2Cl35)

(m/z = 164, 4.20 %)

(m/z = 145, 63.99 %) (m/z = 292, 11 %, 2Cl35Cl37)(m/z = 294, 2.2 %, 2Cl37)

(m/z = 101, 4.22 %)

(m/z = 75, 22.71 %)

(m/z = 292, 22 %)

(m/z = 308, 2.5 %, 2Cl35)(m/z = 310, 1.1 %, 2Cl35Cl37)

(m/z = 312, 2.2 %, 2Cl37)

(m/z = 174, 17.39 %, 2Cl35)(m/z = 176, 6.12 %, 2Cl35Cl37)(m/z = 178, 1.5 %, 2Cl37)

(m/z = 262, 1 %, 2Cl35

(m/z = 264, 0.7 %, 2Cl35Cl37)(m/z = 266, o.15 %, 2Cl37)

(m/z = 173, 100 %, 2Cl35)(m/z = 175, 64.21 %, 2Cl35Cl37)(m/z = 177, 10.32 %, 2Cl37)

(m/z = 145, 63.99%, 2Cl35)(m/z = 147, 41.25 %, 2Cl35Cl37)

(m/z = 149, 11.58 %, 2Cl37)

Cl

Cl

Cl

Cl

Cl

Cl (147e)

+ .+ . + .

+ .

+ .

+ .

+.

+ .

Scheme 3.5: Mass Fragmentation Pattern of 2′-(3′′,5′′-Dichloro-

benzoylmethyl) benzoic acids 147e

Sodium borohydride reduction of keto-acids 147(a-g) afforded the

corresponding racemic hydroxyl-acids 148(a-g), which were cyclodehydrated

with acetic anhydride to produce (dl)-3-difluorophenyl-3,4-dihydro-

isocoumarins 149(a-d) and (dl)-3-dichlorophenyl-3,4-dihydroisocoumarins

149(e-g) which exhibited the carbonyl absorptions at 1703-1710 cm-1 in IR

spectra. The typical AB pattern for C3-H and ABX pattern for C4-H protons

were observed in 1H NMR spectrum of the compound 149a. Thus, each of the

C4-H showed a doublet of doublet at δ 3.46 & 3.81 ppm and other doublet of

doublet observed at δ 5.92 ppm due to C3-H. The mass spectrum of 149a

71

Page 91: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

and 149e showed molecular ion peak at m/z 260 (4%)(M+) and 294(M+)

respectively. Almost the same is the case with dihydroisocoumarins 149(b-d) and 149(f-g).

O

O1

2

34

56

78

1' 2'

3'

4'5'6'

149a

HH

F

F

Table 3.5: 1H NMR Data of 3-(3′,5′-Difluorophenyl)-3,4-

dihydroisocoumarin (149a)

1H NMR δ (ppm) Carbon

No 3-(3,5-difluorophenyl)-3,4-dihydroisocoumarin

(149a)

3 5.92 (dd, J = 2.95,13.10 Hz)

4a 3.81 (dd, J = 4.09, 13.04 Hz)

4b 3.46 (dd, J = 2.92, 16.37 Hz)

5 7.51 (dd, J = 2.15, 7.68 Hz)

6 7.67(dd, J = 3.37,5.67 Hz)

7 7.28 (m)

8 8.14 (d, J = 7.58 Hz)

2′ 7.12 (m)

3′ ----

4′ 6.90 (m)

5′ ----

6′ 7.12 (m)

72

Page 92: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OO

O

O

O

F

F

O

-CO

-CO

-F

F.

O-

.

OO F

-F

O

CO

O

F

-CO

-

F

-

-CO2

+

++

+

+

+

(m/z = 147, 4 %) (m/z = 119, 23.1 %)

F

F F

F

F

F

(m/z = 118, 100 %)

-HCO

(m/z = 104, 58.3 %)(m/z = 90, 38.21 %)

(m/z = 192, 11.41 %)

F

(m/z = 142, 1.41 %)

(m/z = 260, 5.21 %)

(m/z = 114, 21.7 %)

(m/z = 211, 9.17 %)

(m/z = 141, 12.22 %)

(149a)

.

++ .

+ .

+

.

+ .

.

Scheme 3.6: Mass Fragmentation Pattern of 3-(3′,5′-Difluorophenyl)-

3,4-dihydroisocoumarin 149a

73

Page 93: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

O1

2

34

56

78

1' 2'

3'

4'5'6'

149e

HH

Cl

Cl

Table 3.6: 1H NMR Data of 3-(3′,5′-Dichlorophenyl)-3,4-

dihydroisocoumarin (149e)

1H NMR δ (ppm) Carbon

No 3-(3′,5′-dichlorophenyl)-3,4-dihydroisocoumarin

(149e)

3 5.93 (dd, J = 2.90, 12.03 Hz)

4a 3.29 (dd, J = 2.90,16.36 Hz)

4b 3.09 (dd, J = 12.10, 16.25 Hz)

5 7.58 (dd, J = 2.24, 7.60 Hz)

6 7.66 (m)

7 7.48 (m)

8 8.15 (dd, J = 1.34, 7.6 Hz)

2′ 7.30 (d, J = 2.6 Hz)

3′ ----

4′ 7.44 (d, J = 2.3 Hz)

5′ ----

6′ 7.30 (d, J = 2.6 Hz)

74

Page 94: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OO

O

O

O

Cl

Cl

O

-CO

-CO

-Cl

Cl.

O-

.

OO Cl

-Cl

O

CO

O

Cl

-CO

-

Cl

-

-CO2

+

++

+

+

+

(m/z = 147, 4 %) (m/z = 119, 23.1 %)

(m/z = 292, 32.55 %, 2Cl35)(m/z = 294, 21.41 %, 2Cl35Cl37)

(m/z = 296, 10.15 %, 2Cl37)

Cl

Cl Cl

Cl

Cl Cl

(m/z = 227, 5.5 %, Cl35)(m/z = 229, 2.41 %, Cl37)

(m/z = 173,5.5.%, 2Cl35)(m/z = 175, 4.9%, 2Cl35Cl37)

(m/z = 177, 5.2 %, 2Cl37)

(m/z = 174, 8.0 %, 2Cl35)(m/z = 176 1.41 %, 2Cl35Cl37)

(m/z = 178,8.8%, 2Cl37)

(m/z = 145, 3.55 %, 2Cl35)(m/z = 147, 2.41 %, 2Cl35Cl37)

(m/z = 149, 1.15 %, 2Cl37) (m/z = 118, 100 %)

-HCO

(m/z = 104, 1.41 %)

(m/z = 90, 38.41 %)

(m/z = 192, 11.41 %)

Cl

(149e)

.

.+ .

+ .+

+ .

+ .

Scheme 3.7: Mass Fragmentation Pattern of 3-(3′,5′-Dichlorophenyl)-

3,4-dihydroisocoumarin 149e

3.1.4 Experimental

The difluoro- and dichlorobenzoic acids were purchased from Aldrich

and used without further purification. All reagents and solvents were

commercially available and used as supplied. The petroleum ether used

corresponds to the fraction with a boiling range of 40-80 °C. The melting

points of the compounds were determined in open capillaries using a

Gallenkemp melting point apparatus and are uncorrected. The infrared

75

Page 95: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

spectra were recorded on a Hitachi model 270-50 spectrophotometer as KBr

disks or as neat liquids. 1H NMR (300 MHz) spectra were recorded on a

Bruker AM-400 as a CDCl3 solution using TMS as an internal standard, while

the EI MS were recorded on a MAT-112-S machine.

General procedure for 3-(Difluorophenyl)- 146(a-d) and (Dichlorophenyl)- isocoumarins 146(a-g)

A mixture of difluorobenzoic acid 144(a-d) (63.3 mmol) / dichlorobenzoic

acid 144(e-g) (53 mmol) and thionyl chloride (76.0 mmol & 63 mmol) was

heated for 30 min in the presence of a drop of DMF under reflux. Completion of

the reaction was determined by the stoppage of gas evolution. Removal of

excess of thionyl chloride was carried out under reduced pressure to afford

difluoro- 145(a-d) (62.0 mmol) and dichlorobenzoyl chlorides (52.0 mmole)

145(e-g)

A mixture of unsubstituted homophthalic acid (11.3 mmol/ 9.48 mmol)

and difluoro- (62.0 mmol) 145(a-d) / dichlorobenzoyl chloride (52.0 mmol)

145(e-g) was heated at 200 oC under reflux for four hours. The mixture was

dissolved in ethyl acetate and aqueous solution of sodium carbonate was

added in order to remove the unreacted homophthalic acid. The organic layer

was separated, concentrated and chromatographed on silica gel using pet

ether (40-80 oC fraction) as eluent to afford 3-(difluorophenyl)- 146(a-d) and 3-

(dichlorophenyl)isocoumarins 146(e-g) as solids, which were further purified by

recrystallization from methanol.

3-(3′,5′-Difluorophenyl)isocoumarin (146a): Yield: 80%; m.p.: 149–151°C;

IR (KBr, νmax, cm-1): 2920(C-H),, 1704(C=O), 1569, 1515(C=C arom), 1241(C-

O), 1042(C-F); 1H NMR (CDCl3, δ ppm): 8.31 (1H, d, J = 7.92 Hz, H-8), 7.74

(1H, m, H-6), 7.54 (1H, d, J = 7.40 Hz, H-5), 7.50 (1H, dd, J = 7.88 Hz, H-7),

7.40(2H, dd, J = 1.98, 8.30 Hz, H-2′, 6′), 6.94 (1H, s, H-4), 6.81(1H, m, H-4′);

EIMS (DMF, m/z, %): 258(56.72, M); Anal. Calcd. (%) for C15H8F2O2: C,

69.77; H, 3.12; Found: C, 69.73; H, 3.10.

76

Page 96: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

3-(2′,3′-Difluorophenyl)isocoumarin (146b): Yield: 77%; m.p.: 156–158°C;

IR (KBr, νmax, cm-1): 2935(C-H),, 1707(C=O), 1566, 1508 (C=C arom),

1242(C-O), 1141(C-F); 1H NMR (CDCl3, δ ppm): 8.31(1H, d, J = 7.90 Hz, H-

8), 7.92(1H, m, H-6), 7.74(1H, m, H-6′), 7.68(1H, d, J = 7.36 Hz, H 5),

7.54(1H, dd, J = 8.2 Hz, H-7), 7.44(1H, m, H-5′), 7.36(1H, m, H-4′), 7.19(1H,

s, H-4); EIMS (DMF, m/z, %): 258(43.60, M). Anal. Calcd. (%) for C15H8F2O2:

C, 69.77; H, 3.12; Found: C, 69.79; H, 3.13.

3-(2′,4′-Difluorophenyl)isocoumarin (146c): Yield; 78%: m.p.: 131–133°C;

IR (KBr, νmax, cm-1): 2927 (C-H), 1709 (C=O), 1562, 1491 (C=C arom), 1248

(C-O), 1141 (C-F); 1H NMR (CDCl3, δ ppm): 8.30 (1H, J = 7.89 Hz, H-8), 7.99

(1H, m, H-6′), 7.73 (1H, m, H-6), 7.53 (1H, d, J = 7.56 Hz, H-5), 7.50 (1H, dd,

J = 3.0, 7.8 Hz, H-7), 7.12 (1H, s, H-4), 6.99 (1H, m, H-5′), 6.93 (1H, m, H-3′);

EIMS (DMF, m/z, %): 258(83, M); Anal. Calcd. (%) for C15H8F2O2: C, 69.77;

H, 3.12; Found: C, 69.72; H, 3.11.

3-(3′,4′-Difluorophenyl)isocoumarin (146d): Yield: 79%; m.p.: 156–159°C;

IR (KBr, νmax, cm-1): 2923 (C-H), 1703 (C=O), 1565, 1511 (C=C arom), 1247

(C-O), 1189 (C-F); 1H NMR (CDCl3, δ ppm): 8.30 (1H, dd, J = 8.0 Hz, H-8),

7.75 (1H, d, J = 1.24, 7.8, 11.2 Hz, H-6′), 7.74 (1H, m, H-6), 7.72 (1H, m, H-

5′), 7.53(1H, dd, J = 1.0, 7.8 Hz, H-5), 7.51 (1H, m, H-2′), 7.23 (1H, m), 6.88

(1H, s, H-4); EIMS (DMF, m/z, %): 258 (77, M). Anal. Calcd. (%) for

C15H8Cl2O2: C, 69.77; H, 3.12; Found: C, 69.71; H, 3.13.

3-(3′,5′-Dichlorophenyl)isocoumarin (146e): Yield: 81%: m.p: 208-210°C;

IR (KBr, νmax, cm-1): 3156 (C-H), 2935 (C-H), 1708 (C=O), 1590, 1516 (C=C

arom), 1099 (C-Cl); 1H NMR (CDCl3, δ ppm): 8.31 (1H, dd, J = 1.9, 7.9 Hz, H-

8), 7.75 (2H, t, J = 1.2 Hz, H-2′, 6′), 7.55 (1H, dd, J = 2.2, 7.6 Hz, H-5), 7.53

(1H, m, H-6), 7.50 (1H, m, H-7), 7.39 (1H, t, J = 1.2 Hz, H-4′), 6.95 (1H , s ,

H-4); EIMS (DMF, m/z, %): 290 (100, M), 292 (67.5, M+2), 294 (13, M+4);

Anal. Calcd. (%) for C15H8Cl2O2: C, 62.07; H, 2.76; Found: C, 61.83; H, 2.73.

77

Page 97: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

3-(2′, 3′-Dichlorophenyl)isocoumarin (146f): Yield: 80%; m.p: 180-182°C;

IR (KBr, νmax, cm-1): 3152 (C-H), 2931 (Ar-H), 1710 (C=O), 1593, 1490 (C=C

arom), 1096 (C-Cl); 1H NMR (CDCl3, δ ppm): 8.33 (1H, dd, J = 2.1, 7.9 Hz, H-

8), 7.74 (1H, m, H-6), 7.56 (1H, dd, J = 2.2, 8.0 Hz, H-5), 7.54 (1H, m, H-7),

7.50 (2H, t, J = 7.8 Hz, H-6′), 7.33 (2H, t, J = 7.8 Hz, H-5′), 7.29(1H, dd, J =

1.9, 7.9 Hz, H-4′), 6.89(1H, s, H-4); EIMS (DMF, m/z, %): 290 (28, M), 292

(17, M+2), 294 (4, M+4); Anal. Calcd. (%) for C15H8F2O2: C, 62.07; H, 2.76;

Found; C, 61.99; H, 2.69.

3-(2′, 5′-Dichlorophenyl)isocoumarin (146g): Yield: 81%; m.p:187-188°C;

IR (KBr, νmax, cm-1): 3155 (C-H), 2935 (Ar-H), 1710 (C=O), 1593 (C=C arom),

1093 (C-Cl); 1H NMR (CDCl3, δ ppm): 8.32 (1H, dd, J = 2.7, 7.9 Hz, H-8), 7.75

(1H, m, H-6), 7.72 (1H, d, J = 2.0 Hz, H-6’), 7.56 (1H, m, H-7), 7.50 (1H, dd, J

= 2.1, 7.8 Hz, H-5), 7.41 (1H, d, J = 8.5 Hz, H-3’), 7.32 (1H, dd, J = 2.4, 8.5

Hz, H-4'), 7.01 (1H, s, H-4); EIMS (DMF, m/z, %): 290 (100, M), 292 (70,

M+2), 294 (13, M+4); Anal. Calcd. (%) for C15H8F2O2: C, 62.07; H, 2.76;

Found; C, 61.89; H, 2.71.

General procedure for 2-(Difluoro/Dichlorobenzoylmethyl)benzoic acid 147(a-g)

A solution of isocoumarins 146(a-g) in ethanol (50 mL) and 5%

potassium hydroxide (100 mL) were refluxed for 4 h. Ethanol was removed

from the reaction mixture by distillation. Ice cold water (20 mL) was added

and the reaction mixture was acidified with hydrochloric acid. The reaction

mixtures were then extracted with dichloromethane (3 × 20 mL). The extracts

were dried (Na2SO4) and evaporated to yield crude solids 147(a-c), which

were recrystallized from methanol.

2′-(3′′,5′′-Difluorobenzoylmethyl)benzoic acid (147a): Yield, 80%; m.p.:

124–126°C; IR (KBr, νmax, cm-1): 3150 (sp2 CH), 2920 (sp3 C-H) , 1708(C=O),

1470 (C-H), 1142 (C-F); 1H NMR (D2O, δ ppm): 8.08 (1H, d, J = 7.23 Hz, H-6),

78

Page 98: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

7.77 (1H, m, H-5), 7.51 (2H, m, H-2′′,6′′), 7.37 (1H, dd, J = 7.6 Hz, H-4), 7.23

(1H, d, J = 7.4 Hz, H-3), 6.98 (1H, dd, J = 8.25, 16.6 Hz, H-4′′), 4.61 (1H, s,

H-1′); EIMS (DMF, m/z, %): 276 (2, M). Anal. Calcd. for C15H10F2O3: C, 65.22;

H, 3.65; Found: C, 65.24; H, 3.66.

2′-(2′′,3′′-Difluorobenzoylmethyl)benzoic acid (147b): Yield, 70%; m.p.:

151–154°C; IR (KBr, νmax, cm-1): 3300–3250 (-OH), 2920, 2835 (sp3 C-H),

1704 (C=O), 1471 (C-H), 1145 (C-F); 1H NMR (D2O, δ ppm): 8.05 (1H, d, J =

7.2 Hz, H-6), 7.73 (1H, m, H-4), 7.51 (2H, m, H-6′′), 7.31 (1H, dd, J = 7.6 Hz,

H-5), 7.27 (1H, d, J = 7.1 Hz, H-3), 6.98 (1H, dd, J = 8.0, 16.6 Hz, H- 4′′,5′′),

4.65 (1H, s, H-1′); EIMS (DMF, m/z, %): 276 (1.5, M). Anal. Calcd. for

C15H10F2O3: C, 65.22; H, 3.65; Found: C, 65.29; H, 3.62.

2′-(2′′,4′′-Difluorobenzoylmethyl)benzoic acid (147c): Yield, 77%; m.p.:

174–177°C; IR (KBr, νmax, cm-1): 3300–3250 (-OH), 2924, 2831(sp3 C-H),

1708 (C=O), 1477 (C-H), 1140 (C-F); 1H NMR(D2O, δ ppm): 8.10 (1H, d, J =

7.25 Hz, H-6), 7.71 (1H, m, H-4), 7.51 (1H, m, H-6′′), 7.35 (1H, dd, J = 7.6 Hz,

H-5), 7.23 (1H, d, J = 7.4 Hz, H-3), 6.97 (1H, dd, J = 8.23, 16.6 Hz, H-5′′),

6.79 (1H, m, H-3′′), 4.62 (1H, s, H-1′); EIMS (DMF, m/z, %): 276 (3.0, M).

Anal. Calcd. for C15H10F2O3: C, 65.22; H, 3.65; Found: C, 65.19; H, 3.67.

2′-(3′′,4′′-Difluorobenzoylmethyl)benzoic acid (147d): Yield, 69%; m.p.:

132–135°C; IR (KBr, νmax, cm-1): 3300–3250 (-OH), 2927, 2837 (sp3 C-H),

1710 (C=O), 1473 (C-H), 1141 (C-F); 1H NMR (D2O, δ ppm): 8.03 (1H, d, J =

7.21 Hz, H-6), 7.73 (1H, m, H-4), 7.55 (2H, m, H-2′′,6′′), 7.33 (1H, dd, J = 7.1

Hz, H-5), 7.24 (1H, d, J = 7.1 Hz, H-3), 6.89 (1H, dd, J = 8.22, 16.1 Hz, H-5′′),

4.62 (1H, s, H-1′); EIMS (DMF, m/z, %): 276 (2.1, M). Anal. Calcd. for

C15H10F2O3: C, 65.22; H, 3.65; Found: C, 65.22; H, 3.69.

2′-(3′′,5′′-Dichlorobenzoylmethyl) benzoic acid (147e): Yield: 81%;

m.p.:187-188°C; IR (KBr, νmax, cm-1): 3300-3250 (-OH), 3155 (C-H), 2935 (Ar-

H), 1710 (C=O), 1593 (C=C arom), 1093 (C-Cl); 1H NMR (D2O, δ ppm): 8.15

79

Page 99: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

(1H, dd, J = 1.2, 7.9 Hz, H-6), 7.94 (2H, d, J = 1.7 Hz, H-2′′and 6′′), 7.56 (1H,

dd, J = 2.4 Hz, H-4′′ ), 7.52 (1H, m, H-5), 7.24 (1H, m, H-4), 7.18 (1H, dd, J

1.5, 7.9 Hz, H-3), 4.62 (2H, s, H-1′); EIMS (DMF, m/z, %): 308 (2.1, M), 310

(1.0, M+2), 312 (1.2, M+4); Anal. Calcd. (%) for C15H10Cl2O3: C, 58.44; H,

3.25; Cl, 25.26; Found; C, 58.23; H, 3.28.

2′-(2′′,3′′-Dichlorobenzoylmethyl)benzoic acid (147f): Yield: 77%; m.p.;

160-162°C; IR (KBr, νmax, cm-1): 3300-3250 (-OH), 3151 (C-H), 2935 (Ar-H),

1712 (C=O), 1590 (C=C arom), 1091 (C-Cl); 1H NMR (D2O, δ ppm): 8.02 (1H,

dd, J = 2.2, 8.0 Hz, H-6), 7.81 (1H, dd, J = 1.4, 7.7 Hz, H-6′′), 7.63 (1H, m,

H-5), 7.42 (1H, t, J = 8.1 Hz, H-4′′), 7.29 (1H, m, H-4), 7.20 (1H, dd, J = 1.5,

8.0 Hz, H-3), 7.06 (1H, t, J = 7.9 Hz, H-5′′), 4.64 (1H, s, H-1’); EIMS (DMF,

m/z, %): 308 (1.7, M), 310 (5.5, M+2), 312 (1.3, M+4); Anal. Calcd. (%) for

C15H10Cl2O3: C, 58.44; H, 3.25; Found: C, 58.30; H, 3.23.

2′-(2′′,5′′-Dichlorobenzoylmethyl) benzoic acid (147g):Yield: 71%;

m.p.:140-142°C; IR (KBr, νmax, cm-1): 3300-3250 (-OH), 3157 (C-H), 2933 (Ar-

H), 1711 (C=O), 1590 (C=C arom), 1094 (C-Cl); 1H NMR (D2O, δ ppm): 8.17

(1H, dd, J = 1.7, 7.9 Hz, H-6), 7.97 (1H, d, J = 1.8 Hz, H-6′′), 7.58 ( 1H, m, H-

5), 7.44 (1H, d, J = 8.8 Hz, H-4′′), 7.41 (1H, d, J = 9.0 Hz, H-3′′), 7.34 (1H,

dd, J = 2.0, 8.0 Hz, H-3), 7.32 (1H, m, H-4), 4.63 (1H, s, H-1’); EIMS (DMF,

m/z, %): 308 (3, M), 310 (2, M+2), 312 (3, M+4); Anal. Calcd. (%) for

C15H10Cl2O3: C, 58.44; H, 3.25; Found: C, 58.33; H, 3.20.

General procedure for 3-(Difluoro/Dichlorophenyl)-3,4-dihydro- isocoumarin 149(a–g)

To a solution of the keto-acids 147(a–d) (2.07mmol) dissolved in 1%

potassium hydroxide solution (25mL), sodium borohydride (0.25g) was added

and the reaction mixture was stirred for 1 h at room temperature. After

acidification with hydrochloric acid, the reaction mixture was extracted with

ethyl acetate (2 × 50mL). The usual workup gave the crude hydroxy-acids

148(a–d), which were dissolved in acetic anhydride (1mL) and heated under

80

Page 100: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

reflux for 2 h. The reaction mixture was cooled, water (25mL) was added and

the reaction mixture was stirred overnight. The crystals that deposited were

collected by filtration and the filtrate was extracted with dichloromethane (2 ×

20mL). The solvent was removed under reduced pressure. The crude

dihydroisocoumarin 149(a–d) was purified by column chromatography on

silica gel using petroleum ether as an eluent.

2-[2′-Hydroxy-2′-(3′′,5′′-difluorophenyl)ethyl]benzoic acid (148a): Yield:

69%; m.p.: 132-133°C; IR (KBr, νmax, cm-1): 3300-3250 (O-H), 2920 (C-H),

1708 (C=O), 1470 (C-H), 1142 (C-F); 1H NMR (D2O, δ ppm): 8.08 (1H, dd, J

= 7.23 Hz, H-6), 7.37 (1H, dd, J = 7.6 Hz,H-5), 7.23 (1H, d, J = 7.4 Hz, H-3),

6.91 (1H, m , J = 8.2 , 16.6 Hz, H-2′′, 4′′,6′′), 4.49 (1H, dd, J = 7.1, 14.2 Hz,

H-2′), 2.67 (1H, dd, J = 6.2, 15.6 Hz, H-1′a), 2.33 (1H, dd, J = 8.2, 15.6 Hz, H-

1′b); EIMS (DMF, m/z, %): 278 (4, M). Anal. Calcd. for C15H12F2O3: C, 64.75;

H, 4.35 ; Found: C, 64.24; H, 4.66.

2-[2′-Hydroxy-2′-(2′′,3′′-difluorophenyl)ethyl]benzoic acid (148b): Yield:

75%; m.p.: 144-146°C; IR (KBr, νmax, cm-1): 3300-3250 (-OH), 2922 (C-H),

1701 (C=O), 1475 (C-H), 1141 (C-F); 1H NMR (D2O, δ ppm): 8.03 (1H, dd, J

= 7.2 Hz, H-6), 7.67 (m, J = 1.6, 7.9 Hz, H-4), 7.39(1H, dd, J = 7.6 Hz, H-5),

7.43 (1H, d, J = 7.4 Hz, H-3), 6.99 (1H, m , J = 8.2 , 16.6 Hz, H- H-5′′,6′′),

6.69 (1H, ddd , J = 8.2 , 16.0 Hz, H- H-4′′), 4.62 (1H, dd, J = 7.1, 14.2 Hz, H-

2′), 3.01 (1H, dd, J = 6.1, 15.0 Hz, H-1′a), 2.53 (1H, dd, J = 8.2, 15.1 Hz, H-

1′b); EIMS (DMF, m/z, %): 278 (2, M). Anal. Calcd. for C15H12F2O3: C, 64.75;

H, 4.35 ; Found: C, 64.77; H, 4.46.

2-[2′-Hydroxy-2′-(2′′,4′′-difluorophenyl)ethyl]benzoic acid (148c): Yield,

79%; m.p.: 129-130°C. IR (KBr, νmax, cm-1): 3300-3250 (-OH), 2923 (C-H),

1705 (C=O), 1477 (C-H), 1145 (C-F).1H NMR (D2O, δ ppm): 8.10 (1H, dd, J =

7.23 Hz, H-6), 7.69 (m, J = 1.6, 7.92 Hz, H-4), 7.45 (1H, dd, J = 7.6 Hz,H-5),

7.23 (1H, d, J = 7.4 Hz, H-3), 7.01 (1H, dd , J = 8.25 , 16.66 Hz, H-,6′′), 6.55

(1H, ddd , J = 8.21 , 16.61 Hz, H- H-3′′,5′′), 4.75 (1H, dd, J = 7.10, 14.25 Hz,

81

Page 101: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

H-2′), 2.99 (1H, dd, J = 6.21, 15.56 Hz, H-1′a), 2.57 (1H, dd, J = 8.20, 15.56

Hz, H-1′b); EI MS (DMF, m/z, %): 278.00(1.1. M+); Anal. Calcd. for

C15H12F2O3: C, 64.75; H, 4.35 ; Found: C, 64.67; H, 4.26.

2-[2′-Hydroxy-2′-(3′′, 4′′-difluorophenyl)ethyl]benzoic acid (148d): Yield,

75.25 % ; m.p.: 121-123°C. IR (KBr, νmax, cm-1): 3300-3250 (-OH), 2933 (C-

H), 1703 (C=O), 1467 (C-H), 1143 (C-F).1H NMR (D2O, δ ppm): 8.09 (1H, dd,

J = 7.23 Hz, H-6), 7.61 (m, J = 1.6, 7.90 Hz, H-4), 7.55 (1H, dd, J = 7.6Hz,H-

5), 7.43 (1H, d, J = 7.4 Hz, H-3), 6.87 (1H, dd , J = 8.20 , 16.6 Hz, H-6′′), 6.71

(1H, ddd , J = 8.15 , 16.76 Hz, H- H-2′′,5′′), 4.85 (1H, dd, J = 7.01, 14.22 Hz,

H-2′), 2.89 (1H, dd, J = 6.12, 15.6 Hz, H-1′a), 2.55 (1H, dd, J = 8.02, 15.36

Hz, H-1′b); EI MS (DMF, m/z, %): 278(1.6, M+). Anal. Calcd. for C15H12F2O3:

C, 64.75; H, 4.35 ; Found: C, 64.60; H, 4.28.

2-[2′-Hydroxy-2′-(3′′, 5′′-dichlorophenyl)ethyl]benzoic acid (148e): Yield:

77%; m.p.: 138-140°C; IR (KBr, νmax, cm-1): 3300-3250 (-OH), 3157 (C-H),

2933 (Ar-H), 1711 (C=O), 1590 (C=C arom), 1094 (C-Cl):; 1H NMR (D2O, δ

ppm): 8.16 (1H, dd, J 1.77, 7.9 Hz, H-6), 7.73 (1H, dd, J 1.78, 8.9 Hz, H-3),

7.78 (1H, m, H-4), 7.57 (1H, m, H-5), 7.05 (3H, t, J 1.76 Hz, H-2′′, 4′′ and 6′′),

4.10(1H, dd, J 7.12, 14.20 Hz, H-2’), 2.60 (1H, dd, J 6.24, 15.6 Hz, H-1′a),

2.25 (1H, dd, J 8.22, 15.6 Hz, H-1′b); EI MS (DMF, m/z, %): 310 (2.1, M+),

312 (1.2, M++2), 314 (1.9, M++4) Anal. Calcd. (%) for C15H12Cl2O3: C, 58.06;

H, 3.87; Found: C, 57.91; H, 3.88.

2-[2′-Hydroxy-2′-(2′′, 3′′-dichlorophenyl)ethyl]benzoic acid (148f): Yield:

70 %; m.p.: 150-152°C; IR (KBr, νmax, cm-1): 3300-3250 (-OH), 3159 (C-H),

2936 (Ar-H), 1713 (C=O), 1593 (C=C arom), 1094 (C-Cl): 1H NMR (D2O, δ

ppm): 8.13 (1H, dd, J 2.1, 7.9 Hz, H-6), 7.78 (1H, m, H-4), 7.73 (1H, dd, J

1.87, 8.9 Hz, H-3), 7.57 (1H, m, H-5), 7.63 (1H, dd, J 1.76, 7.78 Hz, H-6′′),

7.59 (1H, t, J 7.72 Hz, H-5′′), 7.55 (1H, dd, J 1.76, 8.1 Hz, H-4′′), 4.17(1H,

dd, J 7.12, 14.20 Hz, H-2’), 2.63 (1H, dd, J 6.24, 15.6 Hz, H-1’a), 2.22 (1H,

dd, J 8.22, 15.6 Hz, H-1’b); EI MS (DMF, m/z, %): 310 (8.6, M+), 312 (6.5,

82

Page 102: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

M++2), 314 (1.4, M++4); Anal. Calcd. (%) for C15H12Cl2O3: C, 58.06; H, 3.87;

Found; C, 58.12; H, 3.77.

2-[2′-Hydroxy-2′-(2′′, 5′′-dichlorophenyl)ethyl]benzoic acid (148g): Yield:

72 %; m.p.: 172-174°C; IR (KBr, νmax, cm-1): 3300-3250 (-OH), 3153 (C-H),

2936 (Ar-H), 1716 (C=O), 1593 (C=C arom), 1099 (C-Cl): 1H NMR (D2O, δ

ppm): 8.15 (1H, dd, J 1.87, 7.79 Hz, H-6), 7.78 (1H, m, H-4), 7.73 (1H, dd, J

2.3, 8.9 Hz, H-3), 7.42 (1H, m, H-5), 7.27 (1H, d, J 1.0 Hz, H-6′′), 7.24 (1H,

dd, J 1.56, 7.8 Hz, H-4′′), 7.23 (1H, d, J 8.9 Hz, H-3′′), 4.14(1H, dd, J7.12,

14.20 Hz, H-2’), 2.61 (1H, dd, J 6.24, 15.6 Hz, H-1’a), 2.21 (1H, dd, J 8.22,

15.6 Hz, H-1’b); EI MS (DMF, m/z, %): 310 (2.3, M+), 312 (5.5, M++2), 314

(1.8, M++4); Anal. Calcd. (%) for C15H12Cl2O3: C, 58.06; H, 3.87; Found: C,

57.99; H, 3.58.

(dl)-3-(3′, 5′-Difluorophenyl)-3,4-dihydroisocoumarin (149a): Yield: 69% ;

m.p.: 121-122°C; IR (KBr, νmax, cm-1): 2920 (C-H), 1704, 1244, 1142 (C-F); 1H

NMR (CDCl3, δ ppm): 8.14 (1H, d, J = 7.5 Hz, H-8), 7.67(1H, dd, J = 3.3, 5.6

Hz, H-6), 7.51 (1H, dd, J = 2.1, 7.6 Hz, H-5), 7.28 (1H, m, J=1.7, 6.7 Hz, H-7),

7.12 (1H, m, H-2′, 6′), 6.90 (1H, m, H-4′), 5.92 (1H, dd, J = 2.9, 13.1 Hz, H-3),

3.81 (1H, dd, J = 4.0, 13.0 Hz, H-4a), 3.46 (1H, dd, J = 2.9, 16.3 Hz, H-4b);

EIMS (DMF, m/z, %): 261 (4, M++1); Anal. Calcd. For C15H10F2O2: C, 69.23;

H, 3.87; Found: C, 69.23; H, 3.03.

(dl)-3-(2′,3′-Difluorophenyl)-3,4-dihydroisocoumarin (149b): Yield: 80%;

m.p.: 118-121°C; IR (KBr, νmax, cm-1): 2935 (C-H), 1707, 1244, 1141(C-F); 1H

NMR (CDCl3, δ ppm): 8.13 (1H, d, J = 7.2 Hz, H-8), 7.68(1H, dd, J = 3.2,

5.7Hz, H-6), 7.62 (1H, dd, J = 2.5, 7.5 Hz, H-5), 7.54 (1H, dd, J = 2.2, 7.9 Hz,

H-7), 7.46 (1H, m, J = 5.0, 8.6 Hz, H-5′), 6.71 (m, J = 2.3, 8.5 Hz, H-6′), 6.61

(1H, m, H-4′), 5.76 (1H, dd, J = 2.9, 12.1 Hz, H-3), 3.31 (1H, dd, J = 4.0, 16.3

Hz, H-4a), 3.09 (1H, dd, J = 2.9, 16.3 Hz, H-4b); EIMS (DMF, m/z, %): 260

(5.1, M+). Anal. Calcd. For C15H10F2O2: C, 69.23; H, 3.87; Found: C, 69.23;

H, 3.03.

83

Page 103: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

(dl)-3-(2′,4′-Difluorophenyl)-3,4-dihydroisocoumarin (149c): Yield: 80%;

m.p.: 121-122°C; IR (KBr, νmax, cm-1): 2935(C-H), 1707, 1244, 1141(C-F); 1H

NMR (CDCl3, δ ppm): 8.13 (1H, d, J = 7.2 Hz, H-8), 7.68(1H, dd, J = 3.2, 5.7

Hz, H-6), 7.62 (1H, dd, J = 2.5, 7.5 Hz, H-5), 7.54 (1H, dd, J = 2.2, 7.9Hz, H-

7), 7.46 (1H, m, J = 5.0, 8.6 Hz, H-5’), 6.71 (m, J = 2.3, 8.5 Hz, H-6′), 6.61

(1H, m, H-3′), 5.76(1H, dd, J = 2.9,12.1 Hz, H-3), 3.31(1H, dd, J = 4.0, 16.3

Hz, H-4a), 3.09 (1H, dd, J = 2.9,16.3Hz, H-4b); EIMS (DMF, m/z, %): 260 (11,

M+). Anal. Calcd. For C15H10F2O2: C, 69.23; H, 3.87; Found: C, 69.23; H,

3.03.

(dl)-3-(3′,4′-Difluorophenyl)-3,4-dihydroisocoumarin (149d): Yield: 69%;

m.p.: 121-122°C; IR (KBr, νmax, cm-1): 2923, 1703, 1243, 1149 (C-F); 1H NMR

(CDCl3, δ ppm): 3.08(1H, dd, J= 3.1, 16.3Hz, H-4b), 3.29(1H, dd, J = 4.4,16.3

Hz, H-4a), 5.46(1H, dd, J = 3.0, 11.9 Hz, H-3), 7.15 (2H, m, J = 7.8, 11.3 Hz,

H-2’, 5’), 7.21(1H, m, J = 3.8, 5.1, 8.2 Hz, H-6’), 7.41 (1H, m, J = 1.1, 8.6 Hz,

H-7), 7.55 (1H, dd, J = 1.2, 7.4 Hz, H-5), 7.88 (1H, m, J = 1.8, 6.9 Hz, H-6),

8.13(1H, d, J = 7.7 Hz, H-8); EIMS (DMF, m/z, %): 260 (9, M+); Anal. Calcd.

For C15H10F2O2: C, 69.23; H, 3.87; Found: C, 69.23; H, 3.03.

(dl)-3-(3′,5′-Dichlorophenyl)-3,4-dihydroisocoumarin (149e): Yield: 73%;

m.p.: 184-186°C; IR (KBr, νmax, cm-1): 3153 (C-H), 2935 (Ar-H), 1708 (C=O),

1593 (C=C arom), 1099 (C-Cl); 1H NMR (CDCl3, δ ppm): 8.15 (1H, dd, J =

1.3, 7.6 Hz, H-8), 7.66 (1H, m, H-6), 7.58 (1H, dd, J = 2.2, 7.6 Hz, H-5), 7.48

(1H, m, H-7), 7.44 (1H, d, J = 2.3 Hz, H-4’), 7.30 (2H, d, J = 2.6 Hz, H-2', 6’),

5.93 (1H, dd, J = 2.9, 12.0 Hz, H-3), 3.29 (1H, dd, J = 2.9, 16.3 Hz, H-4a),

3.09 (1H, dd, J = 12.1, 16.2 Hz, H-4b); EIMS (DMF, m/z, %): 292 (11, M), 294

(10, M+2), 296 (3, M+4); Anal. Calcd. (%) for C15H10Cl2O2: C, 61.64; H, 3.08;

Found: C, 61.44; H, 3.44.

(dl)-3-(2',3'-Dichlorophenyl)-3,4-dihydroisocoumarin (149f): Yield: 77 %;

m.p.: 136-138°C; IR (KBr, νmax, cm-1): 3151 (C-H), 2939 (Ar-H), 1710 (C=O),

1597 (C=C arom), 1097 (C-Cl); 1H NMR (CDCl3, δ ppm): 8.15 (1H, dd, J =

2.6, 7.6 Hz, H-8), 7.66 (1H, dd, J = 2.6, 7.7 Hz, H-5), 7.45 (1H, m, H-7), 7.58

84

Page 104: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

(1H, m, H-6), 7.32 (1H, dd, J = 1.7, 7.9Hz, H-4’), 7.30 (1H, t, J = 5.9 Hz, H-

5’), 7.29 (1H, dd, J = 2.6, 7.4 Hz, H-6’), 5.93 (1H, dd, J = 2.8, 12.0 Hz, H-3),

3.27 (1H, dd, J = 2.9, 16.3 Hz, H-4a), 3.10 (1H, dd, J = 12.1, 16.2 Hz, H-4b);

EIMS (DMF, m/z, %): 292 (58, M), 294 (18, M+2), 296 (13, M+4); Anal. Calcd.

(%) for C15H10Cl2O2: C, 61.64; H, 3.08; Found: C, 61.54; H, 3.04.

(dl)-3-(2',5'-Dichlorophenyl)-3,4-dihydroisocoumarin (149g): Yield: 77 %;

m.p.: 121-124°C; IR (KBr, νmax, cm-1): 3154 (C-H), 2933 (Ar-H), 1712 (C=O),

1598 (C=C arom), 1097 (C-Cl); 1H NMR (CDCl3, δ ppm): 8.15 (1H, J = 1.2,

7.6 Hz, H-8), 7.66 (1H, dd, J = 1.2, 7.7 Hz, H-5), 7.48 (1H, m, H-7), 7.58 (1H,

m, H-6), 7.44 (1H, dd, J = 2.3, 8.1 Hz, H-4’), 7.30 (1H, dd, J = 2.6 Hz, H-6’),

7.24 (1H, dd, J = 7.6 Hz, H-3' ), 5.93 (1H, dd, J = 2.9, 12.0 Hz, H-3), 3.29 (1H,

dd, J = 2.9, 16.3 Hz, H-4a), 3.09 (1H, dd, J = 12.1, 16.2 Hz, H-4b); EIMS

(DMF, m/z, %): 292 (35, M), 294 (21, M+2), 296 (11, M+4); Anal. Calcd. (%)

for C15H10Cl2O2: C, 61.64; H, 3.08; Found: C, 61.44; H, 3.44.

3.2 Synthesis of 3H-Furo[3,4-c]-isochromene-1,5-

dione (152) (an unusal isocoumarin) and 3-(3′,4′,5′-

trimethoxyphenyl)isocoumarin (153)

3.2.1 Synthetic scheme

As a continuation of our previous studies196-197 and biological activities

associated with 3-substituted isocoumarins, prompted us to synthesize some

new 3-substituted isocoumarins. Synthetic scheme is shown as follows

(Scheme 3.8)

85

Page 105: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

O

OH

O

R OH

O

SOCl2R Cl

O

200°C, 6hCl

O+

R

HO

150a) R = CH2Cl

O

O

O

150b) R =

where

O

OO

O

OO

O

O

O

(152)

(153)

150(a-b) 151(a-b)

O

O

Cl[Yield = 66%]

[Yield = 66%]

[Yield = 71%]

Scheme: 3.8

3.2.2 Results and discussion Chloroacetic acid (150) was converted into their respective acid

chlorides (151a) by reaction with thionyl chloride in the presence of a drop of

DMF and direct condensation of acid chloride (151a) with homophthalic acid

at 200oC afforded 3,4-substituted isocoumarins (152). Unfortunately, the

desired 3-chloromethylisocoumarin was not detected and 3H-Furo[3,4-

c]isochromene-1,5-dione (152) formed instead of 3-chloromethylisocoumarin which was characterized by spectral data and X-ray crystallography.

3H-Furo[3,4-c]isochromene-1,5-dione (152) was purified by column

chromatography and showed a single spot on TLC. This isocoumarin (152)

did not exhibit characteristic 1H singlet at δ 6.81-7.19 ppm for C4-H in 1H NMR

which indicates the absence of C4-H but a singlet of two protons in aliphatic

86

Page 106: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

region was observed. The aromatic protons appeared in the acceptable

region i.e. 7.50-8.31 ppm (Table). In IR spectra of isocoumarins (152) lactonic

carbonyl absorptions were observed at 1702 - 1709 cm-1. The molecular ion

peak in the mass spectrum was obtained at m/z 202. The structure was finally

confirmed by X-ray crystallography.

The tentative mechanism for the formation of 3H-Furo[3,4-

c]isochromene-1,5-dione (152) is outlined in scheme 3.9. The reaction

presumably occurs by the initial attack of acid chloride (151a), formed under

the reaction conditions, at the active methylene group of homophthalic acid to

form condensation product (Scheme 3.9) Intermediate A undergoes

cyclization to produce isocoumarin derivative B. Further cyclization of B

followed by water elimination results in the formation of 3H-Furo[3,4-

c]isochromene-1,5-dione (152).

O

OH

O

OH

O

SOCl2Cl

O

200°C, 6hCl

O+

HO

O

OO

O

(152)

Cl Cl

Cl

90°C, 30min

OH

OCOOH

ClO

O

HO O

Cl

OOH....

-H+

O

O OHO-

Cl

O

H+

O

O OHOH

Cl

O

-HCl

-H2O

(A)

(B)

Scheme 3.9: Proposed mechanism for the syntesis of 3H-Furo[3,4-c]-

isochromene-1,5-dione

The detail of X-ray crystallographic data is as follows.

87

Page 107: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

a. X-ray crystal structure of 3H-Furo[3,4-c]-isochromene-1,5-dione

The molecular structure of the title compound is shown in Fig. 3.1. In

the title compound, all bond lengths and angles are within normal ranges. The

five member ring is planar to the isocoumarin ring. The bonds lengths within

phenyl ring lie between 1.386 (4)°A and 1.411 (4)°A which highlights the

aromatic character (Fig. 3.1). In case of five membered ring attached to the

isocoumarin ring, a certain variations are found in bond lengths and bond

angles. These variations are due to presence of O atom on C10 and due to

C10---C11. There is no hydrogen bonding below 3.0°A found in the crystal

lattice.

Fig 3.1: Molecular Structure of the Compound 3H-Furo[3,4-c]-isochromene-

1,5-dione 152

This Part of the chapter has been published: Ghulam Qadeer, Nasim

Hasan Rama, Muhammad Tahir Hussain and Wai-Yeung Wong;

Crystal structure of 3H-Furo [3, 4-c] isochromene-1, 5-dione, Acta

Cryst, 2006, E62, o4022–o4023.

88

Page 108: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Crystal data C11H6O4Mr = 202.16 Triclinic P1 a = 7.9892 (13) °A b = 8.0899 (13) °A c = 8.2075 (13) °A α = 67.089 (3)° β = 83.871 (3)° γ = 62.738 (3)° V = 432.68 (12) °A3

Z = 2 Dx = 1.552 Mg m−3

Dm not measured Mo K_ radiation λ = 0.71073 °A µ = 0.120 mm−1

T = 293 (2) K Block Pale yellow 0.32 × 0.24 × 0.20 mm

Geometric parameters (°A, °) Selected bond lengths C1—C2 1.375 (3) C1—C6 1.389 (2) C5—C6 1.405 (2) C5—C9 1.443 (2) C6—C7 1.476 (2) C7—O1 1.196 (2)

C7—O2 1.405 (2) C8—C9 1.330 (2) C8—O2 1.348 (2) C8—C11 1.484 (3) C9—C10 1.458 (2) C10—O3 1.207 (2)

Selected bond angles

C2—C1—C6 119.60 (15) C3—C4—C5 119.49 (15) C4—C5—C9 125.06 (14) C6—C5—C9 115.88 (14)

C1—C6—C5 120.36 (15) O2—C8—C11 122.09 (15) C8—C9—C10 106.99 (15) C5—C9—C10 132.42 (14)

Selected torsional angles H11A—C11—H11B 109.2 C6—C1—C2—C3 0.1 (3) C3—C4—C5—C6 −0.3 (2)

C6—C5—C9—C8 0.6 (2) C4—C5—C9—C10 0.0 (3)

3,4,5-Trimethoxybenzoic acid was purified by recrystalization before

use and its crystal structure is also reported. The X-ray crystallographic data

of 3,4,5-trimethoxybenzoic acid is as follows.

89

Page 109: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

b. X-ray crystal structure of 3,4,5-Trimethoxybenzoic acid

Fig. 3.2: Molecular Structure of Compound 150b

In the title crystal structure, the bonds lengths within phenyl ring lie

between 1.374(4) %A and 1.399(4) %A which highlights the aromatic

character (Fig. 3.2). The valence angle C3--C2--C7 [121.5(3)%] is larger than

the standard value of 120%. The opening of this angle is due to the presence

of the methoxy and carboxyl groups on C2 and C6, respectively, which

involves a decrease of the ring angles of C7 [119.0(3)\%] and C5 [119.7(2)%].

The C1---O1 [1.264(3) %A] bond distance are compatible with

respective distances in related structures198 and smaller than those usually

observed in carboxylic acids (1.365%A). The three methoxy groups are nearly

coplanar with the benzene ring (C3-C4-O3-C8 = 7.2, C4-C5-O4-C9 = 7.1 and

C7-C6-O5-C10 = 4.5). As observed in 2,6-dimethoxybenzoic acid or 2,6-

dimethoxy-3-nitrobenzoic acid, the hydrogen interaction from the hydroxyl O7

of one molecule to the remote carbonyl O2 of a neighbour results in

90

Page 110: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

catemers. The torsion angle between the plane of the acid group and the

benzene ring (C3-C2-C1-O2) is 176.0 (3). The crystal structure is stablized by

inter and intra-molecular hydrogen bonding (Fig. 3.3)

Fig 3.3: A partial packing diagram of 150b, hydrogen bonds are shown as

dashed lines

Crystal data C10H12O5 F000 = 448 Mr = 212.20 Dx = 1.389 Mg m−3

Monoclinic, Pc Mo Kα radiation λ = 0.71073 Å Hall symbol: P -2yc Cell parameters from 1520 reflections a = 7.3384 (3) Å θ = 2.8–22.6º b = 8.8325 (3) Å µ = 0.11 mm−1

c = 15.7560 (5) Å T = 298 (2) K β = 96.576 (2)º Monoclinic, colorless V = 1014.53 (6) Å 3 0.25 × 0.15 × 0.13 mm Z = 4

This Part of the chapter has been published: Ghulam Qadeer, Nasim

Hasan Rama, Murat Tas¸, Okan Zafer Yesilel and Wai-Yeung Wong;

Crystal structure of 3,4,5-Trimethoxybenzoic acid; Acta Cryst.; 2007,

E63 ,o3456.

91

Page 111: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Geometric parameters (°A, °) Selected bond lengths C1—O1 1.243 (4) C12—C17 1.372 (4) C2—C3 1.373 C16—C17 1.385 (4)

C6—C7 1.392 (4) C17—H17 0.9 C7—H7 0.99 (3) C18—O8 1.420 (4)

Selected bond angles O1—C1—O2 123.1(3) C17—C12—C11 118.7(2) O1—C1—C2 119.8(3) C13—C12—C11 119.4(3) C4—C5—C6 120.4(3) C12—C17—C16 119.4(3) O5—C6—C7 123.9(3) C12—C17—H17 120.3 O5—C6—C5 116.0(2) C16—C17—H17 120.3 Selected torsional angles C3—C4—C5—O4 −177.4 (3) C14—C15—C16—C17 −0.9 (5) O3—C4—C5—C6 179.1 (3) C13—C12—C17—C1 6 0.8 (5) C3—C4—C5—C6 −0.5 (5) O4—C5—C6—O5 −2.0(4) O10—C16—C17—C12 178.8 (3) C15—C16—C17—C12 −0.3 (5) Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A C9—H9A···O9i 0.96 2.53 3.406 (4) 152 C9—H9C···O7ii 0.96 2.49 3.405 (4) 159 C18—H18A···O5iii 0.96 2.51 3.321 (4) 142 C19—H19C···O1iv 0.96 2.50 3.404 (5) 158 O2—H2···O7v 0.82 1.83 2.641 (3) 172 O6—H6···O1vi 0.96 (2) 1.71 (2) 2.656 (3) 170 (5)

Symmetry codes: (i) x, −y, z+1/2; (ii) x−1, y, z; (iii) x, −y+1, z−1/2;

(iv) x+1, y, z; (v) x−1, −y+1, z−1/2; (vi) x+1, −y+1, z+1/2.

3,4,5-Trimethoxybenzoic acid (150b) was converted into their

respective acid chloride (151b) by reaction with thionyl chloride in the

presence of a drop of DMF. Direct condensation of acid chloride (151b) with

simple homophthalic acid at 200oC afforded 3-(3,4,5-trimethoxyphenyl)-

isocoumarin (153). This isocoumarin (153) was purified by column

chromatography and showed a single spot on TLC.

92

Page 112: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

This isocoumarin (153) exhibited characteristic 1H singlet at δ 7.26

ppm for C4-H in 1H NMR. The aromatic proton appeared in the acceptable

region i.e. 7.44-8.30 ppm (Table. 3.7). In IR spectra of isocoumarins (153),

lactonic carbonyl absorptions were observed at 1702 cm-1. The molecular ion

peaks in the mass spectrum of 3-(3,4,5-trimethoxyphenyl)isocoumarin (153)

were obtained at m/z 312. The mass fragmentation pattern is shown in

scheme 3.10.

OO

O

O

O

O

OO

O1

23

4 56

78

9

1011

1 23

456

7

81'

2' 3'

4'5'6'

Table 3.7: 1H NMR spectral data of compounds 152 and 153

Carbon 152 153

4 --- 7.26 (1H, s, H-4)

5 7.54 (1H, d, J = 7.54 Hz) 7.78 (1H, d, J = 7.56 Hz)

6 7.74 (1H, m) 7.69-7.74 (1H, m)

7 7.50 (1H, dd, J = 7.88 Hz) 7.44 (1H, m)

8 8.31 (1H, d, J = 7.82 Hz) 8.30 (1H, J = 7.89 Hz)

9 5.01(2H, s) ---

2′ --- 7.62 (2H, d, J = 1.8 Hz)

6′ --- 7.62 (2H, d, J = 1.8 Hz)

OCH3 --- 3.91-3.94 (9H, s, 3 × OCH3)

The structure of the 3-(3,4,5-trimethoxyphenyl)isocoumarin (153) was

also confirmed by X-ray crystallography. The X-ray crystallographic data is as

follows.

93

Page 113: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

OO

O

O

O

OO

O

O

OO

O

O

O

O

O

O

O

OO

O

OO

O

OO

O(m/z = 312, 100 %) (m/z = 284, 68.8 %)

(m/z = 253, 54.6 %)(m/z = 89, 15.3 %)(m/z = 117, 21.0 %)

(m/z =145, 7.8 %)

(m/z = 167, 19.6 %) (m/z = 195, 19.5 %)

-.

-CO

-.

-CO

-CO

-OCH3.

+

+++

+

+ +

-. -CO

. + .

(153)

Scheme 3.10: Mass Fragmentation Pattern of 3-(3′,4′,5′-trimethoxy-

phenyl)isocoumarin (153) c. X-ray crystal structure of 3-(3,4,5-trimethoxyphenyl)isocoumarin

A perspective view of (I) is shown in Fig 3.4. Bond lengths and angles

can be regarded as normal. The molecules are essentially planar (r.m.s.

deviation for all non H atoms = 0.033 A %). No hydrogen bonding is found

within the crystal lattice.

Crystal data: C18H16O5 F000 = 656 Mr = 312.31 Dx = 1.416 Mg m−3

Monoclinic, P 2(1)/n Melting point: 437(1) K Hall symbol: -P2yn Mo Kα radiation λ = 0.71073

94

Page 114: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

a = 9.1496 (11) Å Cell parameters from 2460 reflections b = 12.8426 (15) Å θ = 2.3–26.3º c = 12.6835 (14) Å µ = 0.10 mm−1

β = 100.592 (2)º T = 100 (2) K V = 1465.0 (3) Å3 Irregular, colourless Z = 4 0.60 × 0.50 × 0.40 mm

Fig 3.4: Molecular Structure of the Compound 153

Geometric parameters (°A, °)

Selected bond lengths C7—C8 1.392 (2) O1—C9 1.391 (2) C7—H7 0.9500 O2—C9 1.209 (2) C12—C13 1.393 (2) O5—C18 1.426 (2)

C2—H2 0.9500 C3—C4 1.405 (2) C3—C8 1.408 (2) C4—H4 0.9500 C5—C6 1.410 (2) C5—H5 0.9500

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Mohammad Azad Malik, Javeed Akhtar and Madeleine Helliwell;

Crystal structure of 3-(3,4,5-Trimethoxyphenyl)-1H-isochromen-1-one; Acta

Cryst. 2007, E63, o3447.

95

Page 115: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

Selected bond angles C13—O4—C17 113.61 (13) C10—C11—H11 120.0 C14—O5—C18 117.34 (13) C12—C13—C14 119.54 (15) C5—C4—H4 119.9

C10—C15—H15 120.1 C3—C4—H4 119.9 C4—C5—C6 120.63 (16) C4—C5—H5 119.7

Selected torsional angles

C9—O1—C1—C2 −1.0 (2) C2—C1—C10—C15 2.5 (3) C4—C5—C6—C7 −0.9 (3) C17—O4—C13—C14 −75.9 (2) C5—C6—C7—C8 −0.9 (3) O3—C12—C13—O4 −4.2 (2) C6—C7—C8—C3 2.3 (3) C2—C3—C8—C9 −3.0(2) 3.2.3 Experimental

The chloroacetic acid and 3,4,5-trimethoxybenzoic acid were

purchased from Aldrich and used with further purification by recrystallization.

All reagents and solvents were commercially available and used as supplied.

The petroleum ether used corresponds to the fraction with a boiling range of

40-80 °C. The melting points of the compounds were determined in open

capillaries using a Gallenkemp melting point apparatus and are uncorrected.

The infrared spectra were recorded on a Hitachi model 270-50

spectrophotometer as KBr disks or as neat liquids. 1H-NMR (300 MHz)

spectra were recorded on a Bruker AM-400 as aCDCl3 solution using TMS as

an internal standard, while the EIMS were recorded on a MAT-112-S

machine. XRD data collection was carried out by Data collection: SMART

(Bruker, 1998); cell refinement: SAINT (Bruker, 1999); data reduction: SAINT;

program(s) used to solve structure: SHELXS97 (Sheldrick, 1997); program(s)

used to refine structure: SHELXL97 (Sheldrick, 1997); molecular graphics:

SHELXTL (Bruker, 1999); software used to prepare material; SHELXTL.

General procedure for 3H-Furo[3,4-c]isochromene-1,5-dione (152) and

3-(3′,4′,5′-Trimethoxyphenyl)isocoumarins (153).

A mixture of chloroacetic acid (150a) (47 mmol) / 3,4,5-

trimethoxybenzoic acid (150b) (28 mmol) and thionyl chloride (39 mmol & 34

96

Page 116: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

mmol) was heated for 30 min in the presence of a drop of DMF under reflux.

Completion of the reaction was determined by the stoppage of gas evolution.

Removal of excess of thionyl chloride was carried out under reduced pressure

to afford chloroacetyl (151a) (46 mmol) and 3,4,5-trimethoxybenzoyl chlorides

(27 mmole) (151b)

A mixture of homophthalic acid (11 mmol/ 7.2 mmol) and chloroacetyl-

(46 mmol) (151a) / 3,4,5-trimethoxybenzoyl chloride (27 mmol) (151b) was

heated at 200 oC under reflux for four hours. The mixture was dissolved in ethyl

acetate and aqueous solution of sodium carbonate was added in order to

remove the unreacted homophthalic acid. The organic layer was separated,

concentrated and chromatographed on silica gel using pet ether (40-80 oC

fraction) as eluent to afford 3H-Furo[3,4-c]isochromene-1,5-dione (152) and 3-

(3′,4′,5′-trimethoxyphenyl) isocoumarin (153) as white crystalline solids, which

were further purified by recrystallization from methanol.

3H-Furo[3,4-c]isochromene-1,5-dione (152): Yield: 66 %; m.p.: 131-132°C;

IR (KBr, νmax, cm-1): 2920 (C-H), 1704 (C=O), 1569(C=C), 1241(C-O),

1142(C-F); 1H NMR (CDCl3, δ ppm ): 8.31 (1H, d, J = 7.92 Hz, H-8), 7.74 (1H,

m, H-6), 7.54 (1H, d, J = 7.4 Hz, H-5), 7.50 (1H, dd, J = 7.8 Hz, H-7), 5.01

(2H, s, H-9); EIMS (DMF, m/z, %): 202 (100, M). Anal. Calcd. (%) for

C11H6O4: C, 65.35; H, 2.99; Found: C, 65.29; H, 3.01.

3-(3′,4′,5′-Trimethoxyphenyl)isocoumarin (153). Yield: 71 %; m.p.: 164-

165°C; (KBr, νmax, cm-1): 2926 (C-H), 1720 (C=O), 1603 (C=C), 1561 (C=C),

1245 (C-O); 1H NMR (CDCl3, δ ppm ): 8.30 (1H, J = 7.8 Hz, H-8), 7.78 (1H, d,

J = 7.5 Hz, H-5), 7.69-7.74 (1H, m, H-6), 7.62 (2H, d, J = 1.8 Hz, H-2′,6′), 7.44

(1H, m, H-7), 7.26 (1H, s, H-4), 3.91 (9H, s, 3 × OCH3); EIMS (DMF, m/z, %):

312 (100, M+); Anal. Calcd. (%) for C18H16O5: C, 69.22; H, 5.16; Found: C,

69.12; H, 5.11.

97

Page 117: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

98

Chapter-4

BIOLOIGICAL ACTIVITIES

Isocoumarins, dihydroisocoumarins and related compounds were tested for the

following activities.

4.1. Antioxidant Studies

4.2. Anti-inflammatory Studies

4.3. Herbicide studies

4.4. Fungicide studies

4.5. Insecticide Studies

4.6. Antifungal studies

4.7. Antibacterial studies

4.8. Brine shrimp lethality (Artemia salina) studies

4.9. Cytotoxicity and Antiviral Activities against different Cell Culture

a. Vero cell culture

b. HeLa cell Culture

c. E6SM cell Culture

4.10. Anti-HIV Studies

4.11. Anti-HBV studies

4.12. Anti-cancer studies

4.13. Antimetastatic studies

4.1 Antioxidant Studies

4.1.1 DPPH radical scavenging assay

Radical scavenging activity of compounds against stable DPPH (2,2-

diphenyl-2-picrylhydrazyl hydrate, Sigma–Aldrich Chemie, Steinheim, Germany)

was determined spectrophotometrically. When DPPH reacts with an antioxidant

compound, which can donate hydrogen, it is reduced. The changes in colour

Page 118: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

99

(from deep-violet to light-yellow) were measured at 515 nm on a UV–Vis light

spectrophotometer (Spectronic Genesys 8, Rochester, USA). Radical

scavenging activity of compounds was measured by slightly modified method of

Brand-Williams as described below. Compound solutions were prepared by

dissolving an appropriate amount of each compound in ethanol. The solution of

DPPH in ethanol was prepared just before UV measurements. 1.5 mL of this

solution was mixed with 0.5 mL of compound solutions in 1 dm path length

disposable microcuvettes (final mass ratio of compound with DPPH was

approximately 3:1). The samples (at 1, 10, 100, 500 and 1000 mM) were kept in

the dark for 30 min at room temperature and then the decrease in absorption was

measured. For the control absorption of blank sample containing the same

amount of ethanol and DPPH solution was measured. The experiment was

carried out in triplicate. Radical scavenging activity was calculated by the

following formula:

AB-AA

AB

% inhibition =

Where

AB is the absorption of blank sample (t = 0 min);

AA is the absorption of tested extract solution (t = 15 min).

4.1.2 Procedure

The DPPH (2,2-diphenyl-2-picrylhydrazyl hydrate, Sigma-Aldrich Chemie,

Steinheim, Germany) radical scavenging effect was evaluated according to basic

method. Ethanol solution (0.5 mL) of various sample concentration (1, 10, 100,

500 and 1000 µM) was added to 1.5 mL DPPH ethanol solution (66.66 µM) 199.

After mixing gently and leaving for 30 min at room temperature in ambar vials,

the absorbance was measured at 515 nm using a spectrophotometer200-201.

Values IC50 of inhibition by all compounds was determined by comparison with a

control group. The antioxidant activity of each sample was expressed in terms of

Page 119: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

100

IC50 (µM) required to inhibit DPPH radical formation by 50% and calculated from

the log-dose inhibition curve.

Table 4.1: Antioxidant activities of isocoumarins 146(a–g), keto-acids 147(a–g), hydroxy-acids 148(a–g) and 3,4-dihydroisocoumarins 149(a–g)

Compound 1µM 10µM 100µM 500µM 1000µM IC50 144e 7.281 22.056 52.248 49.471 51.586 93.3 144f 11.563 25.910 51.820 50.106 51.797 93.3 144g 6.747 30.120 44.578 49.683 51.586 563.4 146a 13.735 8.675 22.169 29.175 30.655 > 1000 146b -4.819 14.458 13.494 15.011 15.011 > 1000 146c -0.917 0.917 12.156 38.991 44.954 > 1000 146d 9.639 16.386 19.759 23.890 42.918 > 1000 146e 16.488 13.919 13.919 11.628 14.588 > 1000 146f 9.639 16.386 19.759 23.890 42.918 > 1000 146g 4.096 12.771 44.096 50.529 52.431 462.6 147a 6.210 26.124 50.749 50.106 53.277 97 147b 16.060 13.919 44.754 54.123 55.603 322.4 147c 4.795 27.837 44.754 50.529 52.008 463.3 147d 5.060 18.795 44.096 46.101 51.374 862 147e 6.638 13.490 50.964 50.529 53.700 97.5 147f 16.060 21.842 52.034 50.951 52.008 94.5 147g 6.747 30.120 44.578 49.683 51.586 563.4 148a 16.060 21.842 52.034 50.951 52.008 94.5 148b 6.638 13.490 50.964 50.529 53.700 97.5 148c 4.096 12.771 44.096 50.529 52.431 462.6 148d 4.795 27.837 44.754 50.529 52.008 433.3 148e 11.349 20.557 50.107 50.529 52.008 99.5 148f 3.614 8.434 45.301 53.066 53.066 337.2 148g 5.060 18.795 44.096 46.101 51.374 862 149a -4.587 5.275 38.991 45.872 45.872 > 1000 149b -0.917 0.917 12.156 38.991 44.954 > 1000 149c -3.211 5.734 8.716 34.633 49.312 > 1000 149d 34.699 17.349 34.940 12.474 15.645 > 1000 149e 7.066 10.278 7.281 0.000 0.211 > 1000 149f 10.361 2.410 -2.892 13.108 15.645 > 1000 149g 34.699 17.349 34.940 12.474 15.645 > 1000

Quercetin - - - - - 29.4

Page 120: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

101

4.1.3 Discussion

The radical scavenging effects of synthetic compounds against stable free

radical DPPH (2,2-diphenyl-2-picrylhydrazyl hydrate) were measured

spectrophotometrically. Values for their IC50 are shown in Table 4.1. Compounds

144a, 144b (starting acids), 147a, 147e, 147f (keto-acids), 148a, 148b and 148e (hydroxyl-acids) were found to be good radical scavengers with IC50 below 100,

i.e 93.3, 93.3, 97.0, 97.5, 94.5, 94.5, 93.3 and 99.5 µM, respectively. Compound

144a, 144b, 148b was the most effective DPPH scavenger with an IC50 value of

93.3. Isocoumarins 146(a–g) and 3,4-dihydroisocumarins 149(a–g) were

considerably inactive with IC50 values over 1000 µM. R

O

OOH

R

OH

OOH

DPPH. DPPH.

DPPHHDPPHH

R

O.

OOH

R

OH

OOH

.

H. DelocalizationRate determining step

R

OH

OOH

.

DPPH.

DPPH.

R

OH

OOH

Donation of a second hydrogen atom

DimerizationDPPH

Complexation

R

OH

OOH

R

OH

OHO

4DPPH.R

OH

OOH

R

OH

OHO

Scheme 4.1: Proposed mechanism for keto-acid/ hydroxy-acid and DPPH

reaction

Page 121: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

102

The contribution of the halogens in the aromatic ring when they guard a

disposition ortho or meta the scavenging of the free radical was enhance, when

electronic disposition was modified, also the antioxidant activity is modified. Part

of a certain structure and particularly halogens position in the molecule determine

antioxidant properties; these proprieties depend on the ability to donate hydrogen

or electron to a DPPH free radical 202-203.

4.2 Anti-inflammatory Studies

Inflammation is a complex phenomenon involving interrelationships

between humoral or cellular reactions and a number of inflammatory mediators. It

is a usual symptom covering different pathologies, and there are still many

questions to be answered in order to understand the inflammatory process as

well as a need for better-tolerated and more efficient non-steroidal anti-

inflammatory drugs. In the pathways of the inflammatory process, the implication

of free radicals is particularly important. It has also been reported that anti-

inflammatory drugs may be effective in the prevention of free radical-mediated

damage 204.

4.2.1. Experimental animals

Adult male Wistar CD-1 mice with a body weight ranging from 20 to 25 g

were used. All animals had free access to food and water and were kept on a

12/12 h light –dark cycle.

4.2.2. TPA-induced mouse ear edema

Mouse ear edema was evaluated following the protocol previously

described205, using groups of three male CD-1 mice. Edema was induced by

Page 122: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

103

topical application of 2.5 mg per ear of TPA dissolved in EtOH. Solutions of the

compounds (1 mg/ear) and the standard drug indomethacin (1 mg/ear) as

reference, dissolved in different solvents according to their solubility,

respectively, were applied to both sides of the right ear (10 mL each side)

simultaneously with TPA. The ear swelling was measured before TPA application

and 4 h after, and the edema was expressed as the increase in thickness.

4.2.3 Discussion

The isocoumarins 146(a–g), keto-acids 147(a–g), hydroxy-acids 148(a–g) and 3,4-dihydroisocoumarins 149(a–g) were evaluated by measuring their anti-

inflammatory activity against TPA-induced inflammation in mice, and the

inhibitory activities were compared with that of indomethacin, a commercially

available anti-inflammatory drug. At a dose of 1.0 mg/ear, the compounds 144g,

146b, 147d, 147e, 148a and 148e showed good activity while the remaining

compounds showed lower or nearly equal activity compared to that of

indomethacin (91.35% at a dose of 1.0 mg/ear). For anti-inflammatory evaluation,

these compounds did not achieve a structural similar analysis as for the

antioxidant activity, because, in case of anti-inflammatory activity we have other

factors that intervene in the biological response (solubility, absorption,

distribution, etc.). Due to the lack of exact data related to the solubility,

absorption and distribution of each compound tested, it is difficult to clearly show

the structure–activity relationship concerning the anti-inflammatory activity.

However, we can qualitatively evaluate the activity by taking account of these

factors.

This Part of the chapter has been published: Ghulam Qadeer, Nasim H.

Rama, M. L. Garduno-Ramırez. Synthesis and anti-inflammatory activity of

fluorinated isocoumarins and 3,4-dihydroiscoumarins. J. Fluorine Chem.,

2007, 128, 641-6.

Page 123: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

104

Table 4.2: Anti-inflammatory activity of isocoumarins 146(a–g), keto-acids 147(a–g), hydroxy-acids 148(a–g) and 3,4-dihydroisocoumarins 149(a–g)

Compound % of inhibition of

inflammation (1 mg/ear)

Compound

% of inhibition of inflammation

(1 mg/ear)

144e 70 147f 63.4 144f 21.5 147g 81.8 144g 105.2 148a 93.1 146a -12.5* 148b 81.8 146b 103.7 148c -64.9* 146c 25.3 148d 35.1 146d 64.8 148e 93.0 146e 64.8 148f 81.8 146f 30.8 148g -64.8 146g 82.1 149a 88.2 147a 31.3 149b 86.1 147b 85.4 149c -14.8* 147c 81.0 149d -54.7 147d 92.2 149e -54.7* 147e 92.1 149f 29.3

INDOMETACIN 91.4 149g -212.5 * Values with negative sign represent pro-inflammatory activity

The in vivo anti-inflammatory activity of these compounds indicated these

compounds as a possible candidate for the development of new drugs to treat

symptoms associated with inflammatory diseases, such as osteoarthritis and

arteriosclerosis. Further studies on the assessment of the COX-1/COX-2

selectivity index and inhibitory potency are in progress.

4.3 Herbicide Studies

Weeds compete with crops for sunshine, water, nutrients, and physical

space and are thus capable of greatly influencing the growth of crops and

undermining both crop quality and yield. Also, many weeds are the harbor or nest

of pathogens, viruses, and pests, which may result in the occurrence and spread

Page 124: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

105

of plant diseases and insect pests in crops. Herbicides, as the main weed control

tool, play a very important role in modern agriculture. Since the discovery of the

herbicide 2,4-dichlorophenoxyacetic acid (2,4-D) by Zimerman and Hitchcock,

the agrochemical industry has successfully developed a wide array of herbicides

with various chemical structures and modes of action206. However, an inevitable

problem associated with the use of herbicides is the occurrence of herbicide-

resistant weeds9. For example, the widespread use of herbicides, such as

chlorsulfuron, atrazine, diclofop-methyl, and paraquat, has caused herbicide

resistance in many weeds. Therefore, it is necessary to develop efficient

herbicides with novel structures and modes of action to overcome the resistance

of weeds.

4.3.1 Procedure Compound with certain concentration was dissolved in acetone or DMF

and diluted with water, and the solutions were added to certain amount of soil as

soil treatment in order that the concentration of test compound was 750 kg/ha,

after this, the weed seeds were geminated, each pot contains 20-30 seeds and

each experiment triplicated. The plant was cultured in the green house at 25-

27°C. After 4 weeks of growth the fresh weight was measured and the inhibition

percentage was calculated according to the corresponding control. For foliate

spray, after the growth of weed for 2 weeks, 750 kg/ha of solution was sprayed,

and the weeds were cultured for another 2 weeks, again fresh weight was

measured and the inhibition percentage was calculated according to the

corresponding control.

Page 125: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

106

Table 4.3a: Herbicide activity (% inhibition) of the Difluorophenylisocoumarins

146(a–d), keto-acids 147(a–d), hydroxy-acids 148(a–d) and 3,4-

dihydroisocoumarins 149(a–d)

AR BC EC DS Compound

Density g / hect ST FS ST FS ST FS ST FS

146a 750 - - - 2.9 2.43 - - -

146b 750 - - 21.3 6.6 - 4.2 - -

146c 750 - - 14.1 24.0 18.9 21.7 24.5 9.8

146d 750 - - 10.6 51.9 6.1 2.3 - -

147a 750 - - - 2.9 2.4 - - -

147b 750 - - - 22.6 - - - -

147c 750 1.7 20.0 9.2 - 10.81 8.49 4.9 1.9

147d 750 - - 39.3 - 8.2 9.95 - -

148a 750 - - - - - - - -

148b 750 - - - - - - - -

148c 750 - 8.0 19.02 12.7 9.4 6.6 1.64 15.6

148d 750 - 8.0 22.7 16.5 5.4 22.6 18.0 7.8

149a 750 13.0 - 1.12 16.0 3.3 - - -

149b 750 - - - - - 16.6 - -

149c 750 - - - - - - - -

149d 750 13.0 - - - - - - -

Clorsulfuron 750 100 100 100 100 100 100 100 100

ST = Soil treatment FS = Foliar spray

Key: (-); No activity

The weeds used for this experiment were Brassica campestris (BC), Echinochloa crusgalli (EC), Amaranthus retroflexus L (AR) and Digitaria.

sanguinalis(L.)Scop (DS).

Page 126: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

107

Table 4.3b: Herbicide activity (% inhibition) of the Dichlorobenzoic acid 144(e-g), dichlorophenylisocoumarins 146(e–g), keto-acids 147(e–g), hydroxy-acids 148(e-

g) and 3,4-dihydroisocoumarins 149(e–g)

AR BC EC DS Compound

Density g / hect ST FS ST FS ST FS ST FS

144e 750 - - 19.1 12.6 7.9 17.7 - -

144f 750 - 20.3 75.2 21.1 26.4 - - -

144g 750 - - 29.2 35.5 14.4 17.6 3.3 -

146e 750 - - - 26.55 - 20.6 - -

146f 750 15.9 15.1 94.9 47.9 - - - -

146f 375 10.1 - 73.0 30.4 - - - -

146f 188 - - 33.7 19.7 - - - -

146f 94 - - 1.69 1.74 - - - -

146g 750 - - 1.5 8.8 5.7 10.3 - -

147e 750 - - 23.2 2.9 18.4 17.9 - -

147f 750 - - 88.5 88.2 20.9 10.3 - -

147g 750 - - 67.9 33.3 19.6 0.4 - -

148e 750 13.0 - 94.3 35.5 23.7 - 11.1 22.

148f 750 1.4 - 79.7 33.1 19.0 14.1 15.1 -

148g 750 - - 78.2 35.2 - 10.7 - -

149e 750 - - - 19.4 5.6 - 1.3 -

149f 750 - - - 52.8 - - - 21.1

149g 750 - - 19.8 - 7.7 14.5 - -

Clorsulfuron 750 100 100 100 100 100 100 100 100

ST = Soil treatment FS = Foliar spray

Key: (-); No activity

Page 127: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

108

4.3.2 Discussion

From the biological assay results in Table 4.3a and 4.3b, which

summarize the herbicide activity of the target compounds, some showed a

excellent herbicide activity in the pre-emergence treatment (soil treatment) than

post-emergence (foliar treatment). Compounds 144f (2,3-dichlorobenzoic acid),

146f [3-(2′,3′-dichlorophenyl)isocoumarin)], 147(f-g) (dichloro-keto-acids) and

148(e-g) (dichloro-hydroxy-acids) exhibited considerable inhibitory activity

against Brassica campestris as compared to Amaranthus retroflexus L,

Echinochloa crus-galli and Digitaria sanguinalis(L.)Scop in pre-emergence (soil

treatment). Even the inhibitory activity of compounds 146f and 148e reaches

95% at 750 g /hectare. The inhibitory activity of compound 146f has also been

measured at a concentration of 375, 188 and 94 g/hectare. It has been found that

with a decrease in concentration per hectare, the activity decreases. The other

synthesized compounds also show little herbicidal activity. Although we have no

information about the mode of action of these compounds, further research on

the modification of their structure and the mode of action is in progress.

A general overview of herbicide activity for difluoro- and dichlorophenyl-

isocoumarins and related compounds show significant herbicide activity as

compared to difluoroisocoumarin series.

4.6 Fungicide Studies

Over the last two decades, there has been a dramatic increase in the rate

of superficial and invasive fungal infections207-211. Modern agriculture relies on

effective control of fungal diseases to increase crop yield and quality and

consequently increase crop value212. No single fungicide can be used for all

disease situations and the widespread use of fungicides can select for fungicide

resistant pathogens. Therefore, there is need for safer and more cost-effective

Page 128: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

109

fungicides, which are easier to use and provide better performance against

resistant pathogens213.

4.4.1 Procedure

Using fungi growth inhibition method for fungicide activity determination as

described by Fan 214. Compound with 500 µg mL-1 of concentration was dissolved

in water by 0.1mL of assistance of DMF and then 500 µg mL-1 of compound in

agar plate was prepared, the fungi was inoculated and cultured in the culture

tank at 24°C-26°C, the diameter of fungi spread was measured two days later,

growth inhibition was calculated by corresponding control.

Table 4.4a: Fungicidal activity (% inhibition) of the Difluorophenylisocoumarins

146(a–d), keto-acids 147(a–d), hydroxy-acids 148(a–d) and 3,4-

dihydroisocoumarins 149(a–d)

Compound Conc. (µg ml-1) FO CA AS GZ PP PA

146a 50 10.9 4.6 18.2 5.8 55.0 - 146b 50 - 4.6 31.8 34.8 40.0 - 146c 50 - - 18.8 16.0 34.4 - 146d 50 - 20.0 45.5 14.7 38.5 35.3 147a 50 - - - - - - 147b 50 - 4.6 63.6 8.7 52.5 - 147c 50 - - 31.3 4 42.6 - 147d 50 - - 9.1 - 34.6 11.8 148a 50 - - - - - - 148b 50 - - - - - - 148c 50 12.0 20.0 18.8 20 42.6 - 148d 50 12.0 13.3 12.5 24 39.3 - 149a 50 22.3 40.9 27.3 31.9 32.5 - 149b 50 2.3 9.1 22.7 11.6 18.8 - 149c 50 - - - - - - 149d 50 5.1 - 50.0 8.7 50.0 -

Amistar 50 100 100 100 100 100 100 Key: (-); No activity

Page 129: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

110

The isocoumarins, dihydroisocoumarins and related compounds were

tested for their fungicidal bioassay. The results are reported as linear growth

inhibition (LGI) against some plant pathogens, e.g

FO = Fusarium oxysporum CA = Cercospora arachidicola

AS = Alternaria solani GZ = Gibberella zeae

PP = Physalospora piricola PA = Phoma asparagi

Table 4.4b: Fungicidal activity (% inhibition) of the Dichlorobenzoic acid 144(e-g), dichlorophenylisocoumarins 146(e–g), keto-acids 147(e–g), hydroxy-acids 148(e-

g) and 3,4-dihydroisocoumarins 149(e–g)

Key: (-); No activity

4.4.2 Discussion

From the biological assay results in Table 4.4a, which summarize the

fungicidal activity of the difluorophenylisocoumarins 146(a–d), keto-acids 147(a–d), hydroxy-acids 148(a–d) and 3,4-dihydroisocoumarins 149(a–d). The

Compound Concentration(µg mL-1) FO CA AS GZ PP PA

144e 50 8.0 13.6 100 11.6 57.5 - 144e 20 - - 20.0 - 24.1 - 144f 50 - 4.6 100 11.6 41.3 - 144f 20 - - - - 16.7 - 144g 50 - - 54.5 11.6 42.5 - 146e 50 2.3 4.6 36.4 23.2 47.5 - 146f 50 2.2 - - 5.8 48.8 - 146g 50 - 10.0 27.3 - 19.2 - 147e 50 - 20.0 24.2 26.5 38.5 23.5 147f 50 - 4.6 22.7 5.8 25.0 - 147g 50 - 10.0 18.2 - 26.9 - 148e 50 5.1 - 9.1 14.5 43.8 - 148f 50 - 10.0 9.1 - 21.2 - 148g 50 8.0 - 50.0 23.2 48.8 - 149e 50 8.0 - 22.7 5.8 43.8 - 149f 50 19.4 18.2 31.8 40.6 41.3 - 149g 50 - 60.0 21.2 11.8 53.9 35.5

Amistar 50 100 100 100 100 100 100

Page 130: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

111

compounds 146a, 147b and 149d exhibited considerable inhibitory activity

against Alternaria solani and Physalospora piricola. The other synthesized

compounds also show fungicidal activity, but that activity is not significant.

Table 4.4b summarizes the fungicidal activity of the dichlorobenzoic acid

144(e-g), dichlorophenylisocoumarins 146(e–g), keto-acids 147(e–g), hydroxy-

acids 148(e-g) and 3,4-dihydroisocoumarins 149(e–g). The compounds 144(e-g),

148f and 149g exhibited considerable inhibitory activity against Alternaria solani,

Gibberella zeae and Physalospora piricola Even the inhibitory activity of

compounds 144e and 144f reaches 100% at 50µg/ml. The other synthesized

compounds also show fungicidal activity, but that activity is not significant.

4.5 Insecticidal Studies 4.5.1 Procedure

Weighing 10mg sample into a 50mL of glass beaker, then 20mL of

acetone was added, the maize leaf was dipped in the sample solution for 5

seconds, and the leaf was put in a petri dish with 10 cm of diameter to evaporate

all solvents. Nine pieces of maize leaves were cut short and put into the petri

dish of 10 cm diameter containing 10 Mythimna separata with 4 instars, the death

rate of insect was detected 24h and 96h later experiment. For Culex pipiens

pallens larva experiment, the 4 instars of insect was dipped into the water

solution for 24 hours, the death rate of insect was detected.

This Part of the chapter has been published: Ghulam Qadeer, Nasim H. Rama,

Zhi-Jin Fan, Bin Liu and Xiu-Feng Liu. Synthesis, herbicidal, fungicidal and

insecticidal activities of dichlorophenylisocoumarins and 3,4-dihydroiscoumarins.

J. Braz. Chem. Soc. 2007, 18(6), 1176-82

Page 131: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

112

Table 4.5: Insecticidal bioactivity of the Difluorophenyisocoumarins 146(a-d) and

difluorophenyl-3,4-dihydroisocoumarins 149(a-d)

Death rate of Mythimna

separata % Death rate of mosquito

larva % Compd.

Concentration24h and

96h Concentration 24h

Status of Mythimna

separata survive

146a 500µg/mL 0 5µg/mL 0 Survive regularly

146b 500µg/mL 0 5µg/mL 0 Survive regularly

146c 500µg/mL 0 5µg/mL 0 Survive regularly

146d 500µg/mL 0 5µg/mL 0 Survive regularly

149a 500µg/mL 0 5µg/mL 0 Survive regularly

149b 500µg/mL 0 5µg/mL 0 Survive regularly

149c 500µg/mL 0 5µg/mL 0 Survive regularly

149d 500µg/mL 0 5µg/mL 0 Survive regularly

control Acetone 0 Acetone 0 Survive regularly

4.5.2 Discussion

All the synthesized compounds were tested for insecticide activity but

none of the synthesized compounds showed any insecticide effects on the test

insects.

4.6 Antifungal Studies Isocoumarins, dihydroisocoumarins and related compounds were tested by

agar tube dilution method215 for their in vitro antifungal bioassay. Results were

reported as linear growth inhibition (LGI) against some human pathogens, e.g

TL = Trichphyton longifusus CG = Candida glabrata

Page 132: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

113

CA = Candida albicans MC = Microsporum canis

FS = Fusarium solani AF = Aspergillus flavus

Linear Growth Inhibition results of dichlorophenyl- and difluorophenyl-

isocoumarins and their 3,4-dihydroderivatives are given in table 4.6.

Correlation between Structure and Antifungal Activities Biological study of the natural products with medicinally useful

properties and some of their derivatives indicates possible relationship of the

chemical structure and over-all biological behavior of these compounds. In the

following section, results of biological activities of various types of synthesized

compounds e.g. isocoumarins, keto-acids, hydroxy-acids and

dihydroisocoumarins have been discussed in detail.

4.6.1 Isocoumarin 146(a-g) Linear growth inhibition data of isocoumarins 146(a-g) against different

pathogens in antifungal bioassay is depicted in table 4.6. The isocoumarin

(146d) showed significant activity against Trichphyton longifusus (70%). These

Isocoumarins 146(a-g) are found to be active against Trichphyton longifusus, and

Microsporum canis, show a little activity against Fusarium solani but are

completely inactive against Candida albicans, Aspergillus flavus and Candida

glabrata.

4.6.2 Keto- acids 147(a-g) Table 4.6 illustrates comparison of antifungal activities of keto-acids

147(a-g). Keto-acids 147c and 147f showed excellent acitivites [73% (147c) and

75 % (147f)] as compared to standard Miconazole (70%) against Trichphyton

longifusus. keto acids 147d, 147f and 147g also show significant acitivity [75% (147d), 85% (147f), 70% (147g)] agianst Microsporum canis. In general, activity

of the keto-acids is greater as compared to isocoumarin. Dichloroketo-acid

Page 133: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

114

(147f) is more active than difluoro-keto-acid (147c). These keto-acids are also

completely inactive against Candida albicans, Aspergillus flavus and Candida

glabrata.

TABLE 4.6: Antifungal activity of isocoumarins 146(a–g), keto-acids 147(a–g), hydroxy-acids 148(a–g) and 3,4-dihydroisocoumarins 149(a–g) as linear growth

inhibition (%) at 200 µg/ml of madia SDA

Name of Fungi Compound TL CG CA MC FS AF 146a 45 - - - - - 146b 34 - - 55 35 - 146c - - - 50 - - 146d 70 - - 65 - - 146e 65 - - 60 - - 146f 50 - - - 35 - 146g - - - 55 - - 147a 55 - - 65 - - 147b - - - - - - 147c 73 - - 40 - - 147d 65 - - 75 - - 147e - - - 30 - - 147f 75 - - 85 - - 147g 65 - - 70 15 - 148a 65 - - 66 - - 148b 56 - - 75 - - 148c 78 - - 54 - - 148d 43 - - - - - 148e 34 - - 73 - - 148f 26 - - 35 - - 148g 70 - - 80 - - 149a 67 - - 53 - 20 149b 23 - - 20 - 149c 54 - - 46 - - 149d 65 - - 80 - - 149e 35 - - 30 - 25 149f 55 - - 20 20 - 149g 50 - - 40 - 20

Std. Drugs (Miconazole)

Inhibition (% )

70 110.8 110.8 98.4 73.25 Amphotericin

B 20

Key (-) = NO Activity

Page 134: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

115

4.6.3 Hydroxy-acids 148(a-g)

Table 4.6, exhibits results of antifungal activities of hydroxy-acids 148(a-g). Hydroxy-acids 148c and 148g showed excellent activities [78% (148c) and 80

% (148g)] as compared to standard Miconazole (70%) against Trichphyton

longifusus. Hydroxy acids 148b, 148e and 148g also show significant acitivity

[75% (148b), 73% (148e), 80% (148g)] agianst Microsporum canis. In general,

dichlorohydroxy-acids are found to be more active than difluorohydroxy-acids.

These hydroxy-acids are also completely inactive against Candida albicans,

Aspergillus flavus and Candida glabrata. As a whole, these hydroxy-acids are

found less active as compared to keto-acid.

4.6.4 3,4-Dihydroisocoumarins 149(a-g)

Table 4.6 shows comparison of activities of dihydroisocoumarins 149(a-g). Dihydroisocoumarins are less active among the tested isocoumarin, keto-

acids and hydroxyl-acids against Trichphyton longifusus and Microsporum canis.

But dihydroisocoumarins 149a, 149e and 149g showed excellent activity [20% (149a), 25% (149e), 40% (149g)] against Aspergillus flavus while standard drug

inhibition (Amphotericin B) is 20%, whereas isocoumarin, keto-acids and

hydroxy-acids are completely inactive against Aspergillus flavus. These

dihydroisocoumarins show similar behavior against Candida albicans and

Candida glabrata as that of isocomarin, keto-acids and hydroxy-acids.

4.6.5 General Comparison of Antifungal Activities

A general overview upon activities show that some tested compounds

(147c, 147f, 148c, 148g, 149a, 149e, 149g) have shown some higher activity

against some pathogens as compared to standard drugs, however when

comparing activity of these compounds with each other, it can be concluded that

activity of keto-acids is relatively higher than isocoumarin, hydroxy-acids and

dihydroisocoumarins against Trichphyton longifusus and Microsporum canis.

Page 135: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

116

Thus order of antifungal activity of these compounds is

Keto-acids > hydroxy-acids > isocoumarin> dihydroisocoumarins

4.8 Antibacterial Studies

isocoumarins 146(a–g), keto-acids 147(a–g), hydroxy-acids 148(a–g) and 3,4-dihydroisocoumarins 149(a–g) were tested for their anti-bacterial activity

against various bacteria by adopting agar well diffusion method (carron et al).

Bacteria cultures used were

i. Escherichia coli (EC)

ii. Bacillhas subtilis (BS)

iii. Shigella flexenari (SF)

iv. Staphylococcus aureus (SA)

v. Pseudomonas aeruginosa (PA)

vi. Slamonella typhi (ST)

Imipenem was used as standard drug. 24 Hours old culture containing

approximately 104-106 colony forming unit (CFU) was spread on the surface of

muller Hinton agar (MHA) plates. Wells were created in the medium with the help

of a sterile metallic borer. Test samples of different concentrations were added in

their respective wells. Experimental plates were incubated at 37oC for 24 hours

and zones of inhibition were measured and compared with standard drug.

Results are shown in table 4.7.

4.7.1 Discussion

A general overview upon antibacterial activity show that some tested

compounds (146e, 146f, 147e, 147f, 148c, 148d, 148e, 148f, 148g, 149a, 149b,149e, 149g) have shown significant activity against some pathogens but

somewhat lower activity as compared to standard drugs, however when

Page 136: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

117

comparing activity of these compounds with each other, it can be concluded that

all type of compounds i.e. isocoumarin, keto-acides, hydroxy-acids and 3,4-

dihydroisocoumarins show a little activity.

TABLE 4.7: In vitro antibacterial activity of isocoumarins 146(a–g), keto-acids 147(a–g), hydroxy-acids 148(a–g) and 3,4-dihydroisocoumarins 149(a–g) as

linear growth inhibition (%) at 200 µg/ml of madia SDA

Key: (-) = NO Activity

Name of Bacterias Compound EC BS SF SA PA ST 146a 15 12 - 12 - - 146b 14 11 - 15 15 - 146c - - 14 10 - 15 146d - - - - - - 146e 10 21 12 20 - - 146f 24 17 - - 15 - 146g - - 12 10 - - 147a 12 - - - - - 147b - - - - - - 147c 20 - - 10 - - 147d 15 10 17 - 16 12 147e 11 - - 23 - - 147f - - - 9 - 20 147g - - 12 14 - 17 148a 10 - - 16 18 - 148b 07 12 - 15 - - 148c - - 21 14 - - 148d 13 - 12 - 23 - 148e 21 24 - 13 - - 148f 03 - 12 30 - 12 148g 08 - - 20 - - 149a - 22 20 13 11 - 149b 23 - 17 - 20 11 149c 14 12 - 16 - - 149d - - 11 20 - 13 149e 15 - 25 13 - - 149f 15 15 - 20 12 - 149g - 14 13 12 13 11

Std. Drug ( Imipenem) Inhibition

(% ) 30 33 27 33 24 25

Page 137: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

118

4.8 Brine shrimp (Artemia salina) lethality studies

The procedure for the brine shrimp bioassay generally followed the

method developed by Solis et al. (1993) with some modifications. Brine shrimp

eggs (Artemia salina) obtained locally were hatched in artificial sea water using a

petri dish (34 g sea salts/litre deionized water) by incubation under a 60 W lamp,

providing direct light and warmth (24–26°C). Mycotoxin solutions were prepared

in acetonitrile–water (1:1) and microlitre volumes (amounts calculated on the

basis of a final well volume of 200 µl) were transferred to 96-well plates and air-

dried over night. After complete evaporation of the solvent the toxins were

redissolved in 100-µl-sea water. The actual concentrations of the well solutions

were determined by HPLC–MS analyses (quantification based on isotope dilution

with deuterated FB1 as internal standard; Hartl et al., 1999) and considered with

the data evaluation. Each plate included negative controls consisting of 100-µl

acetonitrile–water (1:1) and 200-µl sea water. Diacetoxyscirpenol (DAS) served

as a positive control (40 µl of a 10 µg/ml solution per well). For each toxin three

assays were performed, with six different dose levels and eight replicates per

dose (for NCM-FB1 only one assay was done due to limited amounts of the

compound).

After an incubation time of 24 hr, the hatched nauplii were separated from

the shells and remaining cysts using a Pasteur pipette and transferred to fresh

sea water. This was facilitated by attracting the shrimps to one side of the petri

dish with a light source. 100 µl of this solution containing 10–20 organisms were

pipetted into each well, resulting in a final well volume of 200 µl. To prevent

moisture loss the plates were covered with Parafilm (American National Can,

Greenwich, USA) and then incubated for 48 hr under direct light at 24–26°C and

shaken at 140 rpm. After 2 days of incubation the plates were examined under a

binocular microscope (18-fold magnification) and the numbers of dead (i.e. non-

motile) nauplii in each well were counted. The total numbers of shrimps per well

were determined onto addition of 100-µl acetonitrile.

Page 138: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

119

A general overview upon Brine shrimp (Artemia salina) lethality activity

show that only compound 146c (isocoumarin) show positive lethality and all

remaining compound including keto-acids and hydroxy-acids have no

cytotoxicity.

Table 4.8: Brine shrimp (Artemia salina) lethality bioassay of Difluorophenyl

isocoumarins 146(a-d) and 3,4-dihydroisocoumarins 149(a-d)

Compound Dose (µg/ml)

No of Shrimps

No. of survivors

LD50 (µg/ml)

LD50 (µg/ml) Remarks

100 30 28 10 30 30 146a 1 30 30

-

7.46

No Cytotoxicity

100 30 28 10 30 30 146b 1 30 30

-

7.46 No Cytotoxicity

100 30 1 10 30 19 146c 1 30 29

13.12 7.46 Postive

Lethality

100 30 28 10 30 30 146d 1 30 30

--

7.46 No Cytotoxicity

100 30 28 10 30 30 149a 1 30 30

-

7.46 No Cytotoxicity

100 30 28 10 30 30 149b 1 30 30

-

7.46 No Cytotoxicity

100 30 28 10 30 30 149c

1 30 30

-

7.46 No Cytotoxicity

100 30 29 10 30 30 149d 1 30 30

-

7.46 No Cytotoxicity

Standard drug: Etoposide Key: (-) - No activity

Page 139: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

120

4.9 Antiviral Studies

Antiviral activity against VZV, CMV, HIV-1, HIV-2, vaccinia virus, vesicular

stomatitis virus, Coxsackie virus B4, respiratory syncytial virus, parainfluenza-3

virus, reovirus-1, Sindbis virus and Punta Toro virus was determined essentially

as described previously216-218.

4.9.1 Viruses and cells

The origin of the viruses was as follows: herpes simplex virus type

1 (strain KOS), herpes simplex virus type 2 (strain G) (see reference 8); vaccinia

virus, vesicular stomatitis virus, coxsackievirus type B-4, Sindbis virus, measles

virus, and poliovirus type 1 (see reference 10); reovirus type 1 (ATCC VR-230),

Semliki forest virus (ATCC VR-67), and parainfluenza virus type 3 (ATCC VR-93)

(American Type Culture Collection, Rockville, Md.). The virus stocks were grown

in primary rabbit kidney cells (herpes simplex types 1 and 2, vaccinia virus, and

vesicular stomatitis virus), Vero cells (measles virus, reovirus, coxsackievirus,

and Semliki forest virus), HeLa cells (polio virus), chicken embryo cells (Sindbis

virus), or human embryonic lung cells (parainfluenza virus). The Vero and HeLa

cell lines used in this study were regularly examined for mycoplasma

contamination and found to be mycoplasma free.

4.9.2 Inhibition of virus-induced cytopathogenicity in vitro Confluent cell cultures in microtiter trays were inoculated dilution that

proved infective for 50% of the cell cultures. After 1 h of virus adsorption to the

cells, residual virus was removed and replaced by cell culture medium (Eagle

minimal essential medium) containing 3% fetal calf serum and various

concentrations of the test compounds. Viral cytopathogenicity was recorded as

soon as it reached completion in the untreated virus-infected cell cultures, i.e., at

1 to 2 days for vesicular stomatitis; at 2 days for Semliki forest, coxsackie, and

polio; at 2 to 3 days for vaccinia, herpes simplex types 1 and 2, and Sindbis; and

Page 140: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

121

at 6 to 7 days for reo, parainfluenza, and measles viruses. The antiviral activity of

the compounds is expressed as the concentration required inhibiting viral

cytopathogenicity by 50%.

4.9.3 Cytotoxicity Cytotoxicity measurements were based on two parameters:

(i) Alteration of normal cell morphology and

(ii) Inhibition of host cell macromolecule (DNA, RNA,

and protein) synthesis.

To evaluate cell morphology, confluent cell cultures which had not been

infected but were treated with various concentrations of the test compounds were

incubated in parallel with the virus-infected cell cultures and examined

microscopically at the same time as viral cytopathogenicity was recorded for the

virus-infected cell cultures. A disruption of the cell monolayer, e.g., rounding up

or detachment of the cells, was considered as evidence for cytotoxicity. To

measure inhibition of host cell macromolecule synthesis, the cells were seeded in

Linbro microtiter tray wells (at 300,000 to 400,000 cells per well) in Eagle minimal

essential medium containing 10% fetal calf serum, various concentrations of the

test compounds, and 2.5 µCi of [methyl-3H]thymidine, [5-3H]uridine, or [4,5-

3H]leucine per ml and allowed to proliferate for 16 h at 37°C. The cells were then

treated with 5% ice-cold trichloroacetic acid, washed with 95% ethanol (five

times), air dried and counted for radioactivity in 7.5 ml of Lipoluma scintillation

fluid.

4.9.4 Antiviral activity in vivo

In vivo assays were carried out with both vesicular stomatitis and

coxsackievirus type B-4 in new born (2-day-old) NMRI mice and with vesicular

stomatitis virus in young weaned (25-day-old) NMRI mice. The NMRI mice were

obtained from the Animal Production Center (Proefdierencentrum) of the

Katholieke Universiteit Leuven. Newborn (2-day-old) mice were inoculated

subcutaneously with vesicular stomatitis virus at 4 PFU/0. 1ml per mouse (PFU,

Page 141: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

122

as determined in mouse L-929 cell cultures) or coxsackievirus type B-4 at 5

CCID50 per 0.1 ml per mouse (CCID50, as determined in Vero cells). The mice

then received either a single intra-peritoneal injection of the compound (in 0.1 ml

physiological saline) at 1 h post infection or repeated intra-peritoneal injections of

the compound at 1 h and 1 and 2 days post infection. Young (25-day-old) mice,

weighing 11 to 13 g, were inoculated intranasally with vesicular stomatitis virus at

40 PFU/0.02 ml per mouse and then received either a single intra-peritoneal

injection of the compound (in 0.5 ml of physiological saline) at 1 h post infection

or repeated intra-peritoneal injections of the compound at 1 h and 1, 2, 3, and 4

days post infection.

Table 4.9a: Cytotoxicity and antiviral activities of Dichlorobenzoic acids 144(e-g), dichlorophenylisocoumarins 146(e-g), keto-acids 147(e-g), hydroxy-acid 148(e-

g) and 3,4-dihydroisocoumarins 149(e-g) in HeLa cell Culture

Minimum inhibitory concentrationb (µg/ml)

Compound

Minimum cytotoxic

concentrationa (µg/ml)

Vesicular stomatitis

virus Coxsackie virus B4

Respiratory syncytial

virus 144e 400 >80 (400) >80 (400) >80 144f >400 >400 400 >400 144g ≥400 >400 >400 >400 146e 400 >80 >80 80 146f 80 >16 >16 >16 146g 80 >16 >16 >16 147e ≥3.2 3.2 >3.2 >3.2 147f >400 400 >400 80 147g 400 >80 >80 >80 148e >400 240 >400 >400 148f >400 240 >400 >400 148g >400 >400 >400 >400 149e 400 >80 >80 >80 149f 80 >16 >16 >16 149g 80 >16 >16 >16

Brivudin >400 >400 >400 >400 (S)-DHPA >400 >400 >400 >400 Ribavirin >400 16 240 9.6

Page 142: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

123

Table 4.9b: Cytotoxicity and antiviral activities of Dichlorobenzoic acids 144(e-g), dichlorophenylisocoumarins 146(e-g), keto-acids 147(e-g), hydroxy-acid 148(e-

g) and 3,4-dihydroisocoumarins 149(e-g) in E6SM cell Culture

Minimum inhibitory concentrationb (µg/ml)

Compound Minimum cytotoxic

concentrationa (µg/ml)

Herpes simplex virus-1 (KOS)

Herpes simplex virus-2

(G)

Vaccinia virus

Vesicular stomatitis

virus

Herpes simplex virus-1

TK- KOS ACVr

144e >400 240 240 >400 >400 240 144f >400 400 >400 >400 >400 240 144g >400 400 >400 >400 >400 240 146e ≥80 >80 >80 >80 >80 >80 146f 80 >16 >16 >16 >16 >16 146g ≥1.6 >1.6 >1.6 >1.6 >1.6 >1.6 147e 3.2 >0.64 >0.64 >0.64 >0.64 >0.64 147f ≥400 >400 240 240 >400 >400 147g ≥400 >400 >400 >400 >400 >400 148e 80 >16 >16 >16 >16 >16 148f 80 >16 >16 >16 >16 >16 148g >400 >400 >400 >400 >400 >400 149e 400 >80 >80 >80 >80 >80 149f 400 >80 >80 >80 >80 >80 149g 400 >80 >80 >80 >80 >80

Brivudin >400 0.128 80 3.2 >400 240 Ribavirin >400 240 240 9.6 >400 240 Acyclovir >400 0.0768 0.128 >400 >400 9.6

Ganciclovir >100 0.0192 0.032 >100 >100 0.48

4.9.4 Discussion

The dichlorophenylisocoumarins 146(e-g) and their 3,4-dihydroderivatives

149(e-g) were examined for their antiviral potential in a number of virus assay

systems, each adapted to its optimal cell substrate. In primary rabbit kidney cells

(Table 4.9a), the antiviral assays were performed with herpes simplex virus type

1 and type 2 as well as vaccinia virus and vesicular stomatitis virus as the

challenge viruses. No specific antiviral effects (i.e minimal antivirally effective

Page 143: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

124

concentration ≥ 5-fold lower than minimal cytotoxic concentration) were noted for

any of the compounds against any of the viruses evaluated.

Table 4.9c: Cytotoxicity and antiviral activities of Dichlorobenzoic acids 144(e-g), dichlorophenylisocoumarins 146(e-g), keto-acids 147(e-g), hydroxy-acid 148(e-

g) and 3,4-dihydroisocoumarins 149(e-g) in Vero cell culture

Minimum inhibitory concentrationb (µg/ml)

Compound Minimum cytotoxic

concentrationa (µg/ml)

Para-influenza-

3 virus

Reovirus-1

Sindbis virus

Coxsackievirus B4

Punta Torovirus

144e 400 >80 >80 >80 >80 >80 144f 400 >80 >80 >80 >80 >80 144g 400 >80 >80 >80 >80 >80 146e 80 >16 >16 >16 >16 >16 146f 16 >3.2 >3.2 >3.2 >3.2 >3.2 146g 16 >3.2 >3.2 >3.2 >3.2 >3.2 147e 400 >80 >80 (400) >80 >80 >80 147f ≥400 >400 400 >400 >400 >400 147g 400 >80 >80 >80 >80 (240) >80 148e 400 >80 >80 >80 >80 >80 148f 3.2 >0.64 >0.64 >0.64 >0.64 >0.64 148g 400 >80 >80 >80 >80 >80 149e 3.2 >0.64 >0.64 >0.64 >0.64 >0.64 149f 400 >80 >80 >80 >80 >80 149g 400 >80 >80 >80 >80 >80 144e 80 >16 >16 >16 >16 >16

Brivudin >400 >400 >400 >400 >400 >400 (S)-DHPA >400 >400 400 >400 >400 >400 Ribavirin >400 80 48 >400 >400 16

a Required to cause a microscopically detectable alteration of normal cell morphology b Required to reduce virus induced cytopathogenicity by 50%

4.10 Anti-HIV studies219-225

Anti-HIV activity and cytotoxicity measurement in MT-4 cells were based on

viability of cells222 that had been infected or not infected with HIV and then

Page 144: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

127

exposed to various concentrations of the test compounds. After the MT-4 cell

were allowed to proliferate for 5 days, the number of viable cell was quantified by

a tetrazolium-based colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-

tetrazolium bromide (MTT) method in 96-well microtrays. While the cytopathic

effects of HIV-1 (IIIB) and HIV-2 (ROD) in MT-4 cells was measured by the MTT

procedure24.

4.10.1 Discussion The dichlorophenylisocoumarins 146(e-g) and their 3,4-dihydro-

isocoumarins 149(e-g) were examined for their anti-HIV activity and only some

trace of anti-HIV activity with the dichlorophenyl-3,4-dihyroisocoumarin 149(e-g) was detected. These compounds could be considered as a lead for further

synthesis. One of the first issues to address is to improve the solubility of the

compounds in aqueous medium.

4.11 Anti-HBV Studies

Hepatitis B virus (HBV)-infected hepatitis is one of the most common

infectious diseases in the world. More than 400 million people worldwide are

chronically infected by the hepatitis B virus226. HBV infection causes liver

diseases such as cirrhosis and may be eventually hepatocellular carcinoma.

Current clinical therapies for HBV infections with interferon-a, lamivudine, and

ribavirin have limited efficacy in a significant proportion of patients and often

result in severe side effects227-229. Thus it is urgently needed to develop more

effective and reliable therapeutics for the treatment of HBV infection.

4.11.1 Testing principle: 2.2.15 cells for hepatitis B virus carrier, the

samples inhibition of hepatitis B virus DNA replication and produce HBsAg,

HBeAg capacity.

Page 145: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

128

4.11.2 Testing Materials and Methods Cell lines: 2.2.15 cells; by keeping the room.

Sample: A sample before dissolved in DMSO dubbed appropriate concentration

measured using culture medium for three times diluted, a total of eight dilutions.

Positive control: lamivudine (3TC), Glaxo Wellcome Company.

Major reagents: hepatitis B virus antigen and e antigen s RIA detection kit,

Beijing North Biotechnology Institute; α 32PdCTP, Demopoulos Biological

Engineering Limited.

Test methods : 2.2.15 cell types of 96-well plate, 36 hours after dilution by the

above were added to samples and a positive control, cells were located Kong,

plus 96 hours after the drugs were replaced after dilution containing different

concentrations of the culture fluid samples, Dosing eight days after admission,

respectively Set cell supernatants and 2.2.15 cells, using RIA cell supernatant

detection of HBsAg, HBeAg secretion, dot blot method to detect cell replication of

HBV DNA levels were calculated IC50 and SI.

Table 4.11: Anti-HBV activity of Dichlorophenylisocoumarins 146(e-g), keto-

acids 147(e-g), hydroxy-acid 148(e-g) and 3,4-dihydroisocoumarins 149(e-g)

HBsAg HBeAg DNA

Replication Compounds TC50

µg/ml IC50 µg/ml SI IC50

µg/ml SI IC50 µg/ml SI

146e 8.903 - - - - - - 146f ≥500 - - - - 17.48 3.18 146g ≥500 - - - - - - 147e 38.52 - - - - - - 147f 55.56 - - - - - - 147g 55.56 - - - - 13.57 3.17 148e 240.37 - - - - - - 148f 166.67 - - - - - - 148g 166.67 - - - - 15.23 2.89 149e 96.23 - - - - - - 149f ≥500 - - - - 23.34 5.23 149g ≥500 - - - - - -

lamivudine 1552.12 - - - - 86.59 17.92

Page 146: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

129

4.11.3 Discussion

Dichlorophenylisocoumarins 146(e-g), keto-acids 147(e-g), hydroxy-acid 148(e-g) and 3,4-dihydroisocoumarins 149(e-g) were evaluated for their

cytotoxicities and anti-HBV activities, namely the ability to inhibit the replication of

HBV DNA and the production of HBsAg and HBeAg in HBV-infected 2.2.15 cells.

The results are summarized in Table 4.11. As shown in Table 4.11, none of the

evaluated compounds exhibited inhibitory effects on HBV.

4.12 Anti-Cancer Studies against breast cell (MCF-7) 4.12.1 Cell culture

MCF-7/AZ is a variant of the human mammary carcinoma cell family MCF-

7230. The cells are maintained on tissue culture plastic substrate (Nunc) in a

mixture of Dulbecco’s modified eagle’s medium (DMEM) and HAMF12 (50/50)

(Invtrogen, Carlsbad, CA) supplemented with 250 IU/ml penicillin, 100 µg/ml

streptomycin (Invitrogen) and 10% fetal bovine serum (FBS ) (Invitrogen), at

37°C in a humidified atmosphere containing 10% CO2.

4.12.2 Assay for cell viability

Cell viability was tested in accordance with Romijin et al.231. Briefly,

mitochondrial dehydrogenase activities were measured by an MTT-reagent

(Sigma, St. Louis, MO). Cells were seeded in microtiter plates at an initial density

of 1.5 × 104 cells in 200 µl culture medium and treated with increasing

concentrations of each compound. In each experiment, eight well were used to

determine the mean O.D. referring to cell viability.

4.12.3 Discussion

From the results given in table 4.12 for toxicity against the breast cancer

cells (MCF-7). Compounds 146a, 146c, 147a, 147b and 149d showed activity at

100 - 400 micromolar concentrations.

Page 147: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

130

Table 4.12: Anti-cancer activity against breast cell of Dichlorophenyl-

isocoumarins 146(e-g), keto-acids 147(e-g), hydroxy-acid 148(e-g) and 3,4-

dihydroisocoumarins 149(e-g)

Concentration (microM) Compd. 0 2.5 25 50 100 400 % Viability 100 108 118 112 60 --- 146a

% std 6 2 7 6 --- % Viability 100 120 120 104 90 89 146b

% std 4 5 7 9 2 % Viability 100 97 96 95 94 51 146c % std 7 6 5 6 13 % Viability 100 75 80 82 70 0 146d % std 7 6 11 7 --- % Viability 100 104 90 87 77 48 147a % std 6 2 8 4 8 % Viability 100 92 82 80 80 68 147b % std 5 8 8 4 4 % Viability 100 98 95 91 90 82 147c % std 7 5 7 6 8 % Viability 100 103 98 92 91 89 147d % std 4 8 9 4 8 % Viability 100 93 93 88 80 79 148a % std 7 8 9 6 2 % Viability 100 91 90 89 86 48 148b % std 4 8 4 3 5 % Viability 100 89 86 86 75 71 148c % std 4 9 7 9 8 % Viability 100 100 91 90 85 83 148d % std 8 3 5 7 8 % Viability 100 96 93 82 83 77 149a % std 5 6 7 5 9 % Viability 100 93 93 90 86 85 149b % std 8 4 7 7 7 % Viability 100 75 80 82 83 80 149c % std 7 6 8 5 9 % Viability 100 92 85 80 79 68 149d % std 5 8 8 4 4

Page 148: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

131

4.13 Anti-metastatic Studies 4.13.1 Procedure

This was performed as described previously232. Briefly, six-well plates

were filled with 1.25ml neutralized type I collagen (0.09%)(Upstate

Biotechnology, Lake Placid, NY) and incubated for 1 h at 37°C to allow

gelfication. Non-invasive into collagen type I 233 and served as control for

invasiveness as compared to untreated MCF-7/Az cells. Single cell

suspensions were prepared with trypsin/ EDTA, mixed with the different

compound solutions, seed on top of collagen type I gel and cultured at 37°C

for 24 h. Numbers of cells penetrating into the gel or remaining at the surface

were counted in 12 field of 0.157mm234, using an inverted microscope

controlled by a computer programme. The invasion index expresses the

percentage of invading cells over the total number of cells.

4.13.2 Discussion

From the results given in Table 4.13, for inhibition of invasion of these

cells into collagen, which is an indication of anti-metastatic activity. 3-(3′,4′-

Difluorophenyl)isocoumarin 146d showed excellent activity. It is worth to look

further into possible anti-metastatic properties of this compound.

Page 149: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

132

Table 4.13: Anti-metastatic activity of Dichlorophenylisocoumarins 146(a-d), keto-

acids 147(a-d), hydroxy-acid 148(a-d) and 3,4-dihydroisocoumarins 149(a-d)

Compounds

Collagen type I invasion

Treatment Corresponding OD80

Cells MCF-7/AZ 146a 17 3 146b 16 2 146c 16 2 146d 2 1 147a 15 2 147b 17 2 147c 16 1 147d 16 2 148a 18 3 148b 17 2 148c 18 2 148d 18 3 149a 18 2 149b 16 1 149c 16 2 149d 16 2

+Control 19 1

Page 150: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

146

Chapter-5 INTRODUCTION

Heterocyclic chemistry has now become a separate field of chemistry with

long history, present society and future prospects. The earliest compounds

known to mankind were of heterocyclic origin. Life, like ours, is totally

dependent on the heterocyclic compounds, it takes birth with purine /

pyrimidine bases, nourishes on carbohydrates and in case of disease, is

cured from medicines, many of which are heterocyclic in nature. Today, the

heterocyclic chemistry delivers reagents and synthetic methods of its own

traditional activity in synthesis of drugs, pesticides and detergents as well as

into the related fields such as biochemistry, polymers and material sciences.

5.1 1,2,4-Triazole

The presence of three nitrogen hetero-atoms in five-membered ring

systems defines an interesting class of compounds, the triazole. This may be

of two types, the 1,2,3-triazoles (1) and the 1,2,4-triazoles (2).

NN

NH

NHN

N(2)

NN

NH

NHN

N(1)

The name triazole was first given to the carbon nitrogen ring system

C2N3H3 by Bladin who described its derivatives in early 1885, although the

structures reported slightly incorrect1-2. An alternative name, pyrrodiazole was

given by Andreocci in 1889 regarding it as a member of a class of compounds

analogous to pyrrole.

A little interest emerged in this field from about 1925 to 1946. The

successors of Andreocci carried out most intensive investigations of the

chemistry of 1,2,4-triazoles. The chemical industry got renewed attention in

the synthesis of both simple and fused triazole systems after the discovery

that certain triazoles capable of inhibiting fog formation in photographic

Page 151: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

147

emulsions and some others being useful herbicides and convulsants3. All

triazoles are of synthetic origin and there is no triazole ring system detected

as yet in nature.

5.2 Chemistry of 1,2,4-triazoles

1,2,4-Triazole systems possess some important features.

5.2.1 Aromaticity and stability

The stability of 1,2,4-triazole nucleus is an inherent property of its

aromatic nature. An aromatic sextet is formed by contribution of one π

electron from each atom joined by double bonds and the remaining two

electrons from a nitrogen atom. Such a system is stabilized by resonance and

though the triazole nucleus may be represented by tautomeric forms, each

tautomer is capable of extended resonance and its structure is more correctly

represented as a hybrid to which the following canonical forms contribute3.

NHN

N

NHN

N

NHN

N

NHN

N

NHN

N

NHN

N

It is also necessary to consider the tautomeric form where the imino

hydrogen atom is at the 4-position. The canonical forms that contribute to this

resonance hybrid are given below3.

NN

NH

NN

NH

NN

NH

NN

NH

⋅⋅ --

This representation makes the assumption that the triazole nucleus

actually consists of two hybrid structures, each representing an individual

tautomeric form. In modern theories such a view is incorrect. A more suitable

Page 152: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

148

expression is to regard 1,2,4-triazoles as a true aromatic system, stabilized by

resonance and represented below3.

NN

N

HN

N

N

HN

N

N

H

NN

N

H

It is not intended to represent the charges on a nitrogen atom and on

the hydrogen atom as separate, complete charges but merely as a slight,

overall negative charge on the ring, balanced by a corresponding positive

charge on the hydrogen atom.

5.2.2 Amphoteric nature 1,2,4-Triazoles are amphoteric in nature, forming salts with acids as well as bases. 5.2.3 Tautomerism in triazoles

Tautomerism is possible in both the structural isomers of triazoles.

a. Tautomerism in 1,2,3-triazoles

1,2,3-Triazoles have two tautomeric forms, 1H-1,2,3-triazole (3) and

2H-1,2,3-triazole (4).

N3

N2N1

N3

N2N1

(3) (4)

H H

b. Tautomerism in 1,2,4-triazoles

1,2,4-Triazoles exhibit two tautomeric forms namely [4H]-1,2,4-

triazoles (5) and [1H]-1,2,4-triazoles (6).

N

N

NH

N

HN

N

(5) (6) The higher stability for tautomer (6) is indicated by temperature

coalescene studies, x-rays studies, basicity measurements, dipole moment

studies, NMR-spectra and theoretical methods.

Page 153: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

149

c. Tautomerism in substituted-1,2,4-triazoles

Among the substituted 1,2,4-triazoles, 3-mercapto-1,2,4-triazoles exist

in two tautomeric forms, because the labile hydrogen may be attached either

to the nitrogen or the sulfur atom. It exhibits thione-thiol tautomeric forms

shown below. This compound exists predominantly in thione (7) form4-5.

N NH

N SRH

N N

N SHRH

1 2 21

44

(7) (8)

Chloro-1,2,4-triazoles exist as 3-chloro-1H-1,2,4-triazole (9a), 3-chloro-

4H-1,2,4-triazole (9b) and 5-chloro-1H-1,2,4-triazole (9c). These tautomers

have the stability order; 9a > 9c > 9b according to physical and theoretical

calculations6.

In case of bromo-1,2,4-triazoles, the possible tautomeric forms are, 3-

bromo-1H-1,2,4-triazole (10a), 3-bromo-4H-1,2,4-triazole (10b) and 5-bromo-

1H-1,2,4-triazole (10c). According to physical and theoretical calculations, the

tautomer (10a) and (10c) are of similar energy and the most stable tautomer

is (10c). These calculations agree with the results of Flammang et al 7.

N NH

NX

N N

NXH

N

NX

HN(a) (b) (c)

9) X= Cl10) X = Br

d. Spectroscopic evidence of tautomerism

Generally, the mixtures of tautomers are formed by the compounds

having free NH group. In the dominant isomer, the position of NH proton in

NMR spectra is generally unknown but sometimes spectroscopic comparison

with alkylated compounds is beneficial. It has been shown that alkylation and

Page 154: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

150

acylation of 1,2,4-triazole leads to 1-substituted compounds and in the

absence of other information, the tautomeric mixtures are represented by 1-H

form in both 1,2,3-triazoles and 1,2,4-triazoles8.

5.3 Spectroscopy of 1,2,4-triazole

Ultraviolet, infrared and nuclear magnetic resonance spectroscopic studies are very informative about the structure of 1,2,4-triazoles and their derivatives.

5.3.1 Ultraviolet spectroscopy The unsubstituted 1,2,4-triazole (11) shows a very weak absorption at

205 nm in the ultraviolet absorption spectrum. Bathochromic shift occurs in N-

acetyl-1,2,4-triazole (12), with the absorption band being located at 221.5

nm9. A similar shift in the absorption maximum of 3,5-dimethyl-1,2,4-triazole

(13) appears on conversion into N-acetyl-3,5-dimethyl-1,2,4-triazole (14)10.

N

N

N N

NH

N N

N

N

(12) (13) (14)

N

NH

N

(11)O O

Cyclopentadiene has an absorption maximum at 238.5 nm and by

replacing carbon-carbon unsaturation with carbon-nitrogen unsaturation, a

known hypsochromic shift occurs, therefore, the lower value obtained for

1,2,4-triazoles is understandable11.

A large hyperchromic effect occurs on the acetylation of triazole and its

derivatives which may be compared qualitatively to the similar effect observed

in passing from benzene to acetophenone2.

In case of 5-substituted-3-mercapto-1,2,4-triazoles, the thione-thiol

tautomeric forms can also be differentiated by UV spectroscopy. The

ultraviolet spectra of an ethanolic solution of 5-aryl-3-mercapto-1,2,4-triazoles

usually show two absorption maxima at 252-256 nm and 288-298 nm. The

Page 155: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

151

absorption at 288-298 nm is due to the presence of the chromophoric C=S

group4-5.

5.3.2 Infrared spectroscopy

The infrared spectroscopy is also very useful in characterization of

triazole ring. The absorptions in the region of 1570-1550 cm-1 due to N=N and

in the region of 1640-1560 cm-1 due to C=N functions12 are the diagnostic

features. 4-Amino-1,2,4-triazoles show the characteristic strong N–H

stretching of a primary amine at 3400-3200 cm-1.

In 5-substituted-3-mercapto-1,2,4-triazoles, the thione-thiol tautomeric

forms can also be differentiated in the IR spectra by the presence of C=S

absorption band at about 1325-1300 cm-1 for thione and by characteristic SH

absorption band at about 2600-2550 cm-1 for thiol forms13-14.

The N–H stretching vibrations at 3165 cm-1 and 3450 cm-1 have also

been found supportive of thione-thiol equilibrium. 4-Amino-1,2,4-triazoles

have been characterized by the appearance of N–H bands in the regions of

3200-3100 cm-1. For NH2 group, the absorption bands appear at about 3400-

3300 cm-1.

5.3.3 NMR and mass spectrometry 13C NMR is a powerful tool to characterize 1,2,4-triazol-3-ones. In the

spectrum of 1,2,4-triazol-3-ones two values for chemical shifts are obtained,

one at about 164-173 ppm for imine (C=N) and the other at 150-160 ppm for

carbonyl (C=O) carbon15-16.

In EIMS of 1,2,4-triazoles, a strong molecular ion peak is always

observed and the cleavage of bonds between N1–N2 and N4–C5 has been

observed usually. The triazole ring also undergoes N1–N2 and C3–N4

cleavage17.

Page 156: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

152

5.4 Applications and biological activities

1,2,4-Triazole and its derivatives are an important class of compounds

which possess diverse agricultural, industrial and biological activities18-19,

including anti-microbial 20-21, sedative22-23, anticonvulsant22-23, anticancer 24-25,

anti-inflammatory26, diuretic27-28, antibacterial 29-31, hypoglycemic 32-33,

antitubercular34-35 and antifungal36-37. In recent years, the synthesis of these

heterocyclic compounds has received considerable attention38-43. This wide

range of applications has been covered by more than sixty papers in the

literature, many in the form of patents.

Some important 1,2,4-triazoles along with their applications are as

follows.

NN

NC2H5

NN

N SHCH2PhN

NN

NH

NH

NH

O

Anticonvulsant44 Dopamine-β-hydroxylase Antihypertensive inhibitor 45 activity46

NN

N N N

HN

NH

NN

N

SH

CH3

N

H

C6H5

Insecticide47 Anti-inflammatory48 Antimicrobial49

N N

NHS

NO2 Antiameobic50

5.4.1 Agricultural applications

In the plant protection technology, the research has been promoted to

discover more efficient pesticides to tackle new challenging problems. In order

Page 157: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

153

to selectively control the growth of weeds, a whole range of azole herbicides

has been developed52 exhibiting high levels of activity, application flexibility,

crop tolerance and low levels of toxicity to mammals. Triazoles play an

important role among this classs of heterocycles. A series of 1,2,4-triazole

derivatives have been patented and extensively employed51. One example of

a herbicidal and pesticidal 1,2,4-triazole is given below.

NN

N NH2

O

O

N N

H

N(MeO)(S)PO

(16) (17) Herbicidal52 Pesticidal53-54

5.4.2 Pharmacological applications Over the last few decades, the biological and pharmaceutical properties

of 1,2,4-triazoles have created considerable interest in their synthesis and

chacterization 55-60. 1,2,4-Triazole and its derivatives possess widely differing

activities e.g., bacteriostate 61, bactericide 62, antifungal 63-64, sedative 65, anti-

carcinogen66, tuberculostatic67, anti-inflammatory68,diuretic69, antiviral70,

muscle relaxant71 and antihuman immunodeficiency virus (HIV)72. The

pathogenic fungi cause life threatening infections that have become

increasingly common during the past two decades. Fungal infections are

common in individuals with immuno-compromised hosts, such as patients

undergoing anticancer chemotherapy or organ transplants and patients with

AIDS. Three major fungal infections in immuno-compromised individuals are

candidosis, aspergillosis and cryptococcosis73-74.Whereas the most

widespread human superficial and cutaneous fungal infections are

dermatomycoses such as, toenails and tinea pedis. The common antifungal

agents currently used in clinic are azoles (such as fluconazole, ketoconazole,

and itraconazole), polyenes (such as amphotericin B) nystatin75,

echinocandins (such as caspofungin and micafungin) 76 and allylamines (such

Page 158: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

154

as naftifine and terbinafine) 77. In antifungal chemotherapy, azoles having

fungistatic and broad-spectrum activities are used widely against most yeasts

and filamentous fungi. Flouconazole is preferred as first line antifungal

chemotherapy with relatively low toxicity but is not effective against anvasive

aspergillosis and has suffered severe drug resistance78-79. An improvement of

fluconazole is itraconazole, having a broader antifungal spectrum and better

tolerance but its variable oral absorption and low bioavailability has hampered

its use. The second generation of azoles such as voriconazole80,

posaconazole81 and ravuconazole82, have been developed with improved

profiles. They are noted for their broad antifungal spectrum, low toxicity, and

improved pharmacodynamic profiles.

Glycosylated triazole derivatives like 1-β-D-ribofuranosyl-[1H]-1,2,4-

triazole-3-carboxamide (Virazol)83 belong to the highly potent drugs against

DNA- and RNA-viruses84. Moreover, this compound shows antitumor activity 85 just as the anomeric 1-(2,3,4,6-tetra-O-acetyl-D-glucopyranosyl)-5-nitro-

[1H]-1,2,4-triazole86. The therapeutic use of non-steroidal anti-inflammatory

drugs (NSAIDs) which are used in treatment of a number of arthritic diseases

such as rheumatoid arthritis and osteoarthritis is limited because of their side

effects, such as, gastrointestinal haemorrhage and ulceration87. So, new

drugs having potent anti-inflammatory activity with minimum side effects have

been developed.

5.4.3 Industrial applications

a. Chemical Industry Some selected triazoles have been used as light emitting diodes

(Electroluminescent devices) 88-89. Some triazole systems have extensive use

in the separation of silver from other metal cations in liquid membrane

systems90.

In addition, these compounds are used as synthetic dyes and bleaching

agents91. Moreover, the inks having smooth writing properties also contain

triazole derivatives e.g, 3-amino-5-mercapto-1,2,4-triazole92. These

Page 159: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

155

compounds have also been reported as inhibitors of corrosion of copper,

brass, aluminium and steel in marine environment93 and inhibit fog formation

in photographic emulsions94, plant growth inhibitors95 and herbicides96.

b. Textile industry The triazole derivatives have many applications in textile industry e.g,

sodium salt of a sulphonated triazole derivative possesses good detergent

action and N-benzylated aminotriazoles (18) have useful properties in

inhibiting the acid fading of dyestuff97.

N N

NR

RNH

(18)

c. Cotton industry In the cotton industry, 3-amino-1,2,4-triazole under its trade name

Amizol, has been used as a commercial defoliant for a number of years98.

5.5 Synthetic approaches towards 1,2,4-trizoles The early methods of preparation of 1,2,4-triazoles were simple and low

yields were obtained but they made the nucleus available for study within a

year of the original discovery by Bladin. These have now been replaced by

later modifications99 and by more efficient methods100-102.

5.5.1 From semicarbazides

A method of practical importance involves synthesis of 1-aryltriazoles

from 1-arylsemicarbazides. This is illustrated by the formation of 3-hydroxy-1-

phenyl-1,2,4-1H-triazole (19) from 1-phenylsemicarbazide and boiling

anhydrous formic acid. By heating (19) to over 200˚C with phosphorus

pentasulfide103, 1-phenyl-1,2,4-1H-triazole (20) is obtained in 80% yield.

Page 160: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

156

(19) (20)

HNHN

O

NH2

NH2

HNO

O NNH

N

O

NN

NHO O

HP2S5

200°C

5.5.2 From triazine

The reaction of s-triazine with a substituted hydrazine salt104 gives

substituted 1,2,4-triazoles. For example, from phenyl hydrazine

hydrochloride 1-phenyl-1,2,4-1H-triazole is obtained in 83% yield. The

reaction proceeds by the formation of a substituted formamidrazone as a

result of ring cleavage of s-triazine, which reacts immediately with another

molecule of triazine to yield the substituted triazole.

N

N N 3NH2NHR.HCl HCNNHR

NH2.HCl

3

NN N

N

N

NR

NH4Cl3

R=Phenyl

5.5.3 From thiosemicarbazides

Usually, 1,2,4-triazoles are formed by the cyclization of a preformed

nucleus of the following types:

NCN

CN

CNN

NType A Type B

C

The former type of ring closure is the most efficient method of

synthesis of C-mono-substituted triazoles. In this way, a triazole containing a

mercapto or hydroxyl group is obtained which then may be removed by

oxidation. Thus, the cyclization of 1-acetylthiosemicarbazide with sodium

methoxide in methanol results in the formation of 5-mercapto-3-methyl-1,2,4-

triazole (21) which readily losses the mercapto group on oxidation with nitric

acid105 to form 3-methyl-1,2,4-triazole (22) 106-107.

Page 161: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

157

NHH2N

NH

NN

NH

NN

NH

(21) (22)

ORO

S

HS

It is interesting that 4-benzoyl-1-carbamoyl-3-thiosemicarbazide (23) under various reaction conditions forms different products: with 20% sodium

hydroxide it yields 5-mercapto-3-phenyl-1,2,4-triazole (24) that is normal

cyclization product of 4-acylthiosemicarbazides with alkali; with concenterated

sulfuric acid at room temperature it forms 3-benzamido-5-hydroxy-1,2,4-

triazole (25) by the elimination of hydrogen sulfide; and a hot acetic

anhydride-acetic acid mixture results in the formation of 3-benzamido-5-

methyl-1,2,4-triazole (26)108.

NN

NH

NaOH H2SO4

(CH3CO)2O

CH3COOH

NN

NH

HO(23)(24)

(25)

HS H2N NH

HN

HN

O

S ONH

O

NN

NH

(26)

NHO

Dimova et al.109 synthesized a series of 4-substituted 5-aryl-1,2,4-

triazoles by cyclization of the corresponding substituted thiosemicarbazides.

Nair et al.110 reported an extensive review on the synthesis of 1,2,4-

triazoles and thiazoles from thiosemicarbazide and its derivatives.

5.5.4 From benzalsemicarbazones with ferric chloride

Oxidation of benzalsemicarbazone with ferric chloride solution readily

gives 3-aroyl-5-hydroxy-1,2,4-triazole (27) which is also obtained by the

Page 162: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

158

oxidation of a mixture of benzaldehyde and azodicarbamide (28). The

hydroxyl group is removed on fusion of hydroxyl triazole with phosphorus

pentasulphide forming 3 (or 5)-substituted triazole (29)111.

RHC NH2

HN

N NCRN

NH

FeCl3

125-130°C1 hr

NCRN

NH

P2S5

N N

H2N NH2

130°C1hr

O

HO

FeCl3

OO

O

(27) (28)

(29)

5.5.5 From carboxylic acid hydrazides

The condensation of carboxylic acid hydrazides with carbon disulphide

in ethanolic potassium hydroxide yields potassium 3-aroyldithiocarbazates

(30) that is directly converted to 4-amino-4H-1,2,4-triazole-3-thione (31) with

an excess of hydrazine112. The methylation of (30) with methyl iodide provided

the S-alkylated derivatives (32) that also cyclize to (31) with hydrazine.

RHN

NH2

O

CS2, KOH

C2H5OHR N

H

HN SK

S

O

R NH

HN S

S

O

CH3I

NHN

N SNH2

R

NH2NH2

NH2NH2

(30)

(31) (32)R= C6H11, C6H5, 4-FC6H4

5.5.6 From 1,3,4-oxadiazol-5-thione

1,3,4-Oxadiazol-5-thiones are converted into 4-amino-1,2,4-triazol-5-

thiones with hydrazine hydrate 113-116. Thus, Reid and Heindel114 indicated that

the 5-aryl-2-1,3,4-oxadiazol-5-thione (33) recycled to form 4-amino-1,2,4-

triazole-3-thione (34) with hydrazine hydrate.

Page 163: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

159

NHN

OAr S

NH2NH2NHN

NAr SNH2

(33) (34)

5.5.7 From thiosemicarbazides and carbonyl compounds

The cyclocondensation of 2,4-disubstituted thiosemicarbazides with

carbonyl functions forms triazoles. Reaction of 4-(2-methylallyl)-2-

phenylthiosemicarbazide (35) with ketones in the presence of catalytic

amount of sulfuric acid afforded 3,3-dimethyl-4-(2-methylallyl)-1-phenyl-1,2,4-

triazolidin-5-thione (36) 117.

NRNH2

NHR1

SNHN

N R3SR1

R2COR3

(36)

H2SO4

RR2

(35)

5.5.8 From thiocarbohydrazides and carbohydrazides

The condensation of thiocarbohydrazides (37) with aliphatic and

aromatic carboxylic acids is the choiest method for the preparation of 3-

alkyl/aryl-4-amino-∆2-5-mercapto-1,2,4-triazoline118-121.

The reaction is improved by using carboxylic acids at their melting

points, resulting in the preparation of 3-alkyl / aryl-4-amino-5-mercapto-1,2,4-

triazole (38) 122.

NHNH2

NHNH2

SNN

N RHSNH2

RCOOH

Melting temperature

(38)(37)

The carbohydrazide (39) on treatment with acid forms 4-amino-3-

methyl-∆2-1,2,4-triazolin-5-one (40).

Page 164: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

160

NHNH2

NHNH2

ONHN

NONH2

MeCOOH

(40)(39)

5.5.9 From thiosemicarbazides with benzoyl chloride

Thiosemicarbazide with benzoyl chloride in boiling pyridine or alkali

undergo benzoylation and cyclization resulting in the formation of 4-benzoyl-3-

phenyl-∆2-5-mercapto-1,2,4-triazoline (41)123.

NHNH2

NH2

SNN

N PhHSPhCOCl

(41)

pH>7

O Ph

5.5.10 From phenylthiosemicarbazide with ethylphenyl-

imidate hydrochloride

The reactions of 4-phenylthiosemicarbazide with ethylphenylimidate

hydrochloride illustrated the formation of 3,4-diphenyl-∆2-1,2,4-triazoline-5-

thione (42)124.

NHNH2

NHPhS

NHN

N PhSPh

EtOC(Ph)=NH.HClpH>7

(42)

5.5.11 From condensation of a nitrile and a hydrazide 3,5-Disubstituted 1,2,4-triazoles (43) are synthesized from the

condensation of a nitrile and a hydrazide in a convenient and efficient one

step base catalysed synthesis described by Kap-Sun Yeung and co-workers

from Bristol-Myers Squibb 125. Under the reaction conditions, a diverse range

of functionality and heterocycles are tolerated.

Page 165: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

161

R1 CN H2NNH R2

O K2CO3, C4H9OH

150°C, 1-14hN

N

NHR1

R2R1,R2 = aryl, hetroaryl 12 compounds

(34-83%)(43)

5.5.12 From isothiocyanates Isothiocyanates on condensation with acylhydrazide affords hydrazine

carbothioamides which are cyclized to 3-mercapto-1,2,4-triazoles (44) under

basic conditions126.

(44)

R1N

CS

+R2 N

H

NH2

O

R2 NH

HN

HN

R1

O

SN N

NR2

R1

SHC2H5OH NaOH

The hydrazide derivatives on refluxing with phenylisothiocyanate in

absolute ethanol yield the corresponding phenylthiosemicarbazides (45), which may be cyclized127 in the presence of NaOH.

N N

R1R

NaOHN N

R R1

(45)

HN NH2

O N N

R1R

HN N

HO

NH

SNCS

N NH

N S

5.5.13 From aromatic nitriles Aromatic nitriles (46) on reaction with hydrazine dihydrochloride in the

presence of hydrazine hydrate under microwave irradiation give 3, 5-

disubstituted-4-amino-(4H)-1,2,4-triazoles (47) 128-129.

(46) (47)

N N

NAr

ArNH2

Ar-CN + N2H4.2HClNH2-NH2.H2O

Ethylene glycol

Page 166: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

162

5.5.14 Solid phase synthesis of triazoles

There are, so far, only a few published studies about the solid-phase

synthesis of substituted 1,2,4-triazoles. Katritzky reported the synthesis of tri-

substituted 1,2,4-triazoles on a solid support based on the condensation

reaction between an acyl hydrazide and substituted amidines130. The yields

were 37-90% and the purities depended on the substituents of the triazole

core.

This procedure enables the alkylation of the 1-position, giving the

trisubstituted 1,2,4-triazoles, but suffers from the nontraceless nature of the

reaction sequence. Hence, the synthesized 1,2,4-triazoles contain the 4-

hydroxyphenyl linker of the starting Wang resin. 3,4,5-Trisubstituted 1,2,4-

triazoles were also prepared on solid supports131.

A traceless synthesis of 3,5-disubstituted 1,2,4-triazoles has been

developed on polymeric supports132, using immobilized mesoionic 1,3-

oxazolium 5-oxides (münchnones) as key intermediates in the 1,3-dipolar

cycloaddition reaction, as shown below.

O

O

O OHN

O

OH

O

R

O

N

O

OH

O

R

R1 O

O O

N NN

R1

R

HN NN

R1

R

Page 167: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

163

5.5.15 Synthesis of 1,2,4-triazoles under microwave irradiation

Microwave irradiation has become a widely used method to synthesize

many useful organic chemicals rapidly, with good yields and high

selectivity133-147. A great many relevant works suggest only a thermal nature of

the microwave action, which means that microwaves are considered to be a

method to heat chemical reagents rapidly and without any overheating. Some

other works describe specific non-thermal effects, and these effects are likely

to exist. Sometimes the effects are thought to be only specific forms of heat

effects, but not always.

Kappe et al.148-149 have used this method extensively for the synthesis

of their organic molecules; meanwhile Molteni and Ellis150 reviewed the work

carried out since 1994 in the field of microwave-assisted synthesis of

heterocyclic compounds and reactions in which a heteroatom is directly

participating in the bond forming process that gives rise to a heterocyclic core.

A novel one-step synthesis of thiazolo-[3,2-b]-1,2,4-triazoles (50) was

reported from the reaction of chalcones (48) with bis(1,2,4-triazolyl)sulfoxide

(49)151 . Symmetrical 3,5-substituted 4-amino-1,2,4-triazoles (53) are quickly

prepared from aromatic aldehydes (51) via nitriles (52) by two-step reactions

without any separation under microwave irradiation for each several

minutes152.

R1 R2

O+

N

NN

S NO N

N

Toluene, 90°C-TrH-H2O

NN

N

S

R2

R1

O

(48) (49) (50)

Ar

O

NO

MWNH2OH.HCl

MWNH2NH2.2HClNH2NH2.H2O

HO(CH2)2OH N N

NHHNArAr

N N

NAr

ArNH2

Ar

N

(51) (52) (53)

Page 168: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

164

Condensation of acid hydrazide (54) with S-methylisothioamide

hydroiodide (55) and ammonium acetate on the surface of silica gel under

microwave irradiation afforded 1,2,4-triazoles (56)153.

R1

O

NH

NH2+

R2

S

NH2+ I-

NH2OAc,SiO2

(C2H5)3N, MW N N

HN

R2

R1

(54) (55) (56)

An efficient microwave-assisted one-pot and three-component

synthesis of substituted 1,2,4-triazoles (57) has been achieved utilizing

substituted primary amines154 .

O

NH

NH2 + NO

O+ RNH2

MW

N N

NR

(57)

Kidwai et al.155 have synthesized new antifungal azoles including 1,2,4-

triazole derivatives from substituted hydrazide (58) using various solid

supports under microwave irradiation.

NH

NHRO

NH O

NH4SCN

Basic/ NeutralAlumina, MW N

H O

NH

NNR

S

(58)

A simple and fast synthesis of 6-aryl-3-substituted 5H-[1,2,4]-triazolo-

[4,3-b][1,2,4]-triazoles (60) in high yields has been developed by microwave

assisted heterocyclization of N-(3-methylthio-5-substituted 4H-1,2,4-triazol-4-

yl)benzene- carboximidates (59)156.

Page 169: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

165

HN

NH

NNN

R

S

NN

N

N

NH

R

(59) (60) 5.6 1,3,4-Thiadiazoles

1,3,4-Thiadiazoles (61) are five membered heterocyclic compounds

having two nitrogen and one sulfur atom in a symmetrical structure. The first

representative of this group was discovered by Emil Fischer in 1882. Since

then 1,3,4-thiadiazoles have received a great deal of attention, mainly

because of their diversity of pharmacological properties157.

NN

SR R'

(61) Tautomerism

The substituted 1,3,4-thiadiazoles also exibit tautomerism e.g,

mercapto-substituted thiadiazoles show thiol and thione tautomeric forms. The

tautomerism influences the reactivity of the thiadiazoles, which has been

demonstrated for polymerization processes and substitution reactions at

different moieties158-159.

For 2-mercapto-5-methyl-1,3,4-thiadiazole (McMT), spectral data

indicate that the thione tautomer exists predominantly in DMSO solution as

well as in the solid state.

NHN

S S

NN

S SH

2,5-Dimercapto-1,3,4-thiadiazole, is present as thione-thiol tautomer

(62) in solid state while in solution a solvent dependent equilibrium is believed

to exists between the thione-thiol and thione-thione (63) forms, the former

Page 170: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

166

being the prevailing species in polar solvents shown by vibrational

spectroscopy and X-ray structural analysis160.

NHHN

S SS

NHN

S SHS

NN

S SHHS

(63) (62)

Some important applications of 1,3,4-thiadiazoles are :

NN

S SHHS

NN

S NH2

NN

S SN N

Corrosion inhibitor161 Antitumor and Ulcer Inhibitor163

Neoplasma Inhibitor162

NN

SN Cl

NN

S NHRNN

O

NN

S NH

Insecticidal164 Antibacterial165 H2-antagonist166

5.7 Applications

1,3,4-Thiadiazole derivatives are important in industry, medicine and

agriculture.

a. Biological applications

Thiadiazole ring displays a broad spectrum of biocidal activities

possibly by virtue of NCS toxophoric moiety. Its derivative 2-amino-1,3,4-

thiadiazole is a cyclic analogue of thiosemicarbazone, which often displays

diverse physiological activites. 2,5-Disubstituted-1,3,4-thiadiazole derivatives

have been found to possess biocidal activities including antifungal167-168

Page 171: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

167

antibacterial167-169, anti-inflammatory170, antituberculosis171, anticonvulsant172,

radioprotective173, and anticancer 174.

1,3,4-Thiadiazole derivatives possess interesting biological activity

probably conferred to them by the strong aromaticity of this ring system175,

which leads to great in vivo stability and generally, a lack of toxicity for higher

vertebrates, including humans. When diverse functional groups that interact

with biological receptors are attached to this ring, compounds possessing

oustanding properties are obtained. Except for some antibacterial

sulfonamides (albucid and globucid), no longer used clinically, but which

possessed historical importance 175. The most interesting examples are

constituted by 5-amino-l,3,4-thiadiazole derivatives such as the thiol (64a), a

compound used as radioprotective agent 176, as well as an investigational

antitumor 177 and gastroprotective 178 drug; acetazolamide (64b), which was

the first non-mercurial diuretic drug179-182 used clinically thereafter as

antiglaucoma 183, antiepileptic184 and antiulcer drug 185 together with a large

series of its congeners derived from 5-amino-1,3,4-thiadiazol-2-sulfonamide

(64c) 182.

NN

S ZRHN

64a. R = H, Z= SH64b. R = Ac, Z= SO2NH264c. R = H, Z= SO2NH2

These compounds have also been investigated as complexing

agents180-183 in order to obtain substances with a diversified biological activity,

conferred to them among others by the presence of the metal ions, except for

that of the thiadiazole ligand. Thus, some metal complexes of ligands of type

64a-c have been recently reported as in vitro inhibitors of the zinc enzyme

carbonic anhydrase181-183 whereas in vivo studies showed good antiepileptic

action for some Cu(II) and Zn(II) complexes of the sulfonamide type

ligands182. Finally, some 2,5-disubstituted-l,3,4-thiadiazoles as well as their

Cu(II) complexes were reported to act as fungitoxic agents 183

Page 172: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

168

b) Industrial applications

1,3,4-Thiadiazole, 2,5-bis(tert-nonyldithio) is used to formulate finished

greases and lubricating oils including industrial, gear, automatic transmission

and some types of automotive crankcase, heavy duty diesel and medium

speed diesel oils. In these applications, it is used as an ashless copper

corrosion inhibitor and extreme pressure (EP) agent 183.

1,3,4-Thiadiazole, 2,5-bis(tert-nonyldithio) is also used as a sulfur

deactivator, corrosion inhibitor and antioxidant in gasoline, heating oil and

Liquefied Petroleum Gas 184.

c. Chemical industry

i) Synthesizing other chemicals

1,3,4-Thiadiazoles with amines or alkalis readily produce salts of strong

bases. For example, 2,5-dimercapto-1,3,4-thiadiazole (DMcT), also known as

bismuthiol is used to synthesize salts and polymers. On treatment with

ammonia or pyridine it generates monoammonium and monopyridine salts,

while with hydrazine or hydrazine hydrate gives both mono- and dihydrazine

salts185. Its heavy metal salts may be prepared in a polar solvent such as

ethanol. The products are polymeric complexes186. Polymers may be

produced by treating DMcT with a sulfur chloride such as S2Cl2 in strongly

alkaline solution at temperatures up to 100 °C187. Oxidative polymerization of

2,5-Dimercapto-1,3,4-thiadiazole gives poly(1,3,4-thiadiazole-2,5-diyldithio)

(65).

NN

S SS

n(65)

ii) Flame and scorch retardants

Fire retardant compositions for wildland fire suppression are based on

salts of thiosulfuric acid and contain substituted 1,3,4-thiadizoles presumably

as a stabilizer or the corrosion inhibitor188. Chemonic industries, Inc., USA,

Page 173: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

169

patented colored liquid fire retardant compositions for aerial application to

vegetation. The compositions were apparently based on ammonium

polyphosphate and contained DMcT189. Substituted 1,3,4-thiadizoles or its

sodium or other metal salts were used as a viscosity stabilizer for the

galactomannan gum thickener for a fire retardant composition based on

ammonium phosphate and/or ammonium sulfate190.

iii) Adhesion improver

Substituted 1,3,4-thidizoles salts were used in compositions to

improve heat-resistant adhesion between steel cord and rubber in tires191.

Use of 1,3,4-thidiazole derivtives as the cross linking agent improved

compositions for bonding and sealing chlorinated polyethylene roofing

membranes used for flat roofs 192.

iv) Analytical reagent

Substituted 1,3,4-thidizole is a chelating agent that is used to

determine metals in industrial, environmental, and biological samples (e.g.),

lead193, copper194, aluminum, arsenic, nickel, and selenium 195, and cadmium

and zinc196. Maxwell and Smyth reported satisfactory use in the determination

of cadmium, lead, and zinc in river waters by anodic stripping voltammetry197.

v) Purification and waste treatment

1,3,4-Thidizoles salts and bentonite or zeolite sorbents are used for

heavy metal ion removal from industrial and municipal wastewaters198. A

potential wastewater treatment process is reported for cadmium removal by

complexation and polymerization with derivatives of 1,3,4-thiadizoles199. The

complexing property of DMcT is used to remove trace metal contamination

(cadmium, cobalt, iron (III), lead, nickel, and zinc) from commercial ethanol for

use as engine fuel 200. It has also been employed as a trapping agent for

sulfonic acids from penicillin 201.

Page 174: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

170

vi) Biocides

The biocidal compositions of certain thiol compounds such as 1,3,4-

thiadiazole derivtives in which antimony, arsenic, or bismuth is complexed

with it are prepared229. The biocidal compositions were said to be useful as a

disinfectant, a preservative, bactericidal, bacteriostatic, antibiofilm, antifungal,

and antiviral agents 202.

The thiadiazole fungicides are active against leaf blight of rice and

canker of oranges203.

vii) Photography

DMcT is a general photographic chemical204. DMcT and its disodium

salt were used to form silver salts that stabilized photographic layers205. It (up

to 1%) was used in the composition of a silver halide nuclear emulsion234. The

salts of substituted 1,3,4-thiadizoles are also used in silver-based

photographic materials for metal reliefs, printed circuits, and printing plates 206. Moreover, 1,3,4-thiadizole derivatives are mentioned as a component of

the novel ascorbic acid developer liquid for silver halide film processing in a

more recent Fuji Photo Film Co. patent 207.

5.8 Synthetic approaches towards 1,3,4-thiadiazoles

Several methods for the synthesis of 1,3,4-thiadiazoles are reported in

the literature. Some of them are given below.

5.8.1 From thiosemicarbazide

i) A standard method for the preparation of 1,3,4-thiadiazoles (66) is

dehydrative cyclization of acylthiosemicarbazide

R1 NH

HN R2

O

S

H2SO4 NN

SR1 R2

(66)

Page 175: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

171

Different acidic reagents have been used for dehydration e.g. sulfuric

acid 208, phosphoric acid, acetic anhydride 209-210 and phosphorus halides211.

ii) The condensation of thiosemicarbazide with benzoic acid (67) in

phosphorus oxychloride gives 1,3,4-thiadiazole in 94% yield212.

OH

OH2N N

HNH2

S

NN

S NH2ArPOCl31h, 70°C(67)

iii) 5-Amino-[1,3,4]-thiadiazole derivatives can be prepared from the

reaction of p-anisaldehyde (68) with thiosemicarbazide to give an

intermediate, followed by cyclization in the presence of ferric chloride in

aqueous solution 213

H2N NH

NH2

S

NN

S NH2H3CO

C2H5OH

FeCl3H2O

H3CO

CHON

HN NH2

H3COS

(68)

+

iv) The dehydration of thiosemicarbazides with acetyl chloride followed

by hydrolysis of the acetamide gives amino-1,3,4-thiadiazoles.

NH

HN NH2

O

S

NN

S NH2

C10H21O C10H21O

CH3COClHCl

5.8.2 From diacylhydrazide

2,5-Disubstituted-1,3,4-thiadiazole has been prepared by the reaction

of diacylhydrazide (69) with phosphorus pentasulphide 214.

Page 176: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

172

R1HN N

HR2

O

O

P2S5NN

SR1 R2

+

(69) 5.8.3 From dithiocarbazinic acid derivatives 215

a. Dithiocarbazinic acid derivatives (70) on reaction with carbon

disulphide yield 2, 5- dimercapto-1,3,4-thiadiazole (71).

RHN

NH

SH

S

CS2NN

S SHS

R

+

(70) (71)

b. 2-Mercapto-1,3,4-thiadiazoles can also be obtained when

dithiocarbazinic acids react with aliphatic aldehydes.

RHN

NH

SH

S NN

S CH3HS

RCH3CHO+

5.8.4 From fluorous Lawesson’s reagent

1,3,4-Thiadiazoles are synthesized from N’-acylbenzohydrazide (72) by using Fluorous Lawesson’s reagent (73) in THF at 55°C within 6hrs 216.

NH

HN CH3

O

O

NN

S CH3LR

(72)

P

P

S

S

O(CH2)4(CF2)6F

F(F2C)6(H2C)4O

Fluorous Lawesson's reagent (LR)(73)

Page 177: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

173

5.9 Isatin derivatives-Indolinones 5.9.1 Isatin

Isatin (1H-indole-2,3-dione) was first obtained by Erdman and Laurent

in 1841 as a product from the oxidation of indigo by nitric and chromic acids.

NH

O

O

(74)

The synthetic versatility of isatin has led to the extensive use of this

compound in organic synthesis. Three reviews have been published regarding

the chemistry of this compound: the first by Sumpter, in 1954217, a second by

Popp in 1975218, and the third on the utility of isatin as a precursor for the

synthesis of other heterocyclic compounds219. The synthetic versatility of

isatin has stemmed from the interest in the biological and pharmacological

properties of its derivatives. These properties are more fully detailed in the

supplementary material.

In nature, isatin is found in plants of the genus Isatis220, in Calanthe

discolor LINDL 221 and in Couroupita guianensis Aubl 222, and has also been

found as a component of the secretion from the parotid gland of Bufo frogs223,

and in humans as it is a metabolic derivative of adrenaline224-226. Substituted

isatins are also found in plants, for example the melosatin alkaloids

(methoxyphenylpentyl isatins) obtained from the Caribbean tumorigenic plant

Melochia tomentosa227-229 as well as from fungi: 6-(3’-methylbuten-2’-yl)isatin

was isolated from Streptomyces albus230 and 5-(3’-methylbuten-2’-yl)isatin

from Chaetomium globosum231. Isatin has also been found to be a component

of coal tar232.

Isatin has been known for about 150 years and has been recently

found, like oxindole and endogenous poly-functional heterocyclic compounds,

to exhibit biological activity in mammals233. Isatin also is a synthetically

Page 178: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

174

versatile substrate that can be used to prepare a large variety of heterocyclic

compounds, such as indoles and quinolines, and as a raw material for drug

synthesis 234. Isatin is further known to be a color reagent for the amino acid

proline, forming a blue derivative235. This property has been exploited for the

determination of this amino acid in pollens236 and other vegetable materials237

using paper chromatography or for the detection of polymer-bound

compounds possessing proline residues 238. Some isatin derivatives exhibit

antiplasmodial activity239. Schiff bases and Mannich bases of isatin are known

to possess a wide range of pharmacological properties including antibacterial,

240-242 anticonvulsants, 243-244 anti-HIV245-248, antifungal 249-252 and antiviral

activity253. Bis-Schiff bases are characterized by their capacity to completely

co-ordinate a metal ion, forming chelate rings 254. The Schiff bases of isatin

have also been used as ligands for complexation of metals such as copper

II255. These complexes catalyzed the oxidation of carbohydrates. Bis-Schiff

bases can act as inhibitors of human α-thrombin256. Recently it has been

reported that a bis-imine of isatin has antimicrobial properties 257 and affects

cell viability258.

5.9.2 Indolinones

Oxindoles (2-indolinones) are a class of heterocyclic compounds found

in many natural products259-260 and in a number of marketed drugs261-264. Of

particular interest are 3-substituted 3-hydroxyoxindoles. This substructure is

encountered in a large variety of natural alkaloids with a wide spectrum of

biological activities. Especially 3-alkyl-substituted 3-hydroxyoxindoles occur

frequently in nature, e.g., convolutamydines(75)265, 267, donaxaridine(76) 266-

267, maremycins(77) 268 , dioxibrassinine (78) 269, celogentin K (79) 270 and 3’-

hydroxyglucoisatisins (80) 271.

Page 179: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

175

NH

OBr

BrOH

R

75. Convolutamydine A (R = COCH3)75. Convolutamydine E (R = CH2OH)

N

NH

HN

R

OO

S

HH

77. Maremycin A (R = OH)77. Maremycin B (R = OH)

NH

HOHN

S

S

O

78. Dioxibrassinine

NH

HO

NH

O

76. Danxaridine

NH

HO

O

O

O

NOSO3SO

OH

HO HO

OH

-

80. 3-hydroxyglucoisatisin

NH

O

79. Celogentin K

HO

NH

NHOHN

NH

O

O

OHNO

HNNH2

NHO

HNNH

OCO2H

NHN

5.9.3 Synthetic approaches towards indolinones

The products of partial reduction of isatin, dioxindole and oxindole,

have been widely used in organic synthesis, especially in the development of

new drugs. Some natural products also belong to these classes of

compounds, for instance dioxibrassinin272.

Oxindoles can be prepared by the reduction of either dioxindoles or

isatins. The reductions have been performed by using red phosphorous and

iodic acid273, by use of H2S in a pyridine/co-solvent mixture274. It can also be

prepared by reduction of the isatin-3-ethylene thioketal with Raney nickel275 or

by the Wolf-Kishner reaction276-279. Lower molecular weight alcohols such as

EtOH or iPrOH are used as solvents which lead to high yields of the desired

product280. It has however been found that isatin can be reduced to the

corresponding oxindoles in high yields (76-92%) by the use of hydrazine

hydrate as the solvent in the absence of any additional base281-282. A

chromatographic method for the quality control of oxindoles, frequently used

as raw materials for pharmaceutical products, using normal phase HPLC has

been developed283.

Page 180: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

176

Indigo, isoindigo and indirubin are natural pigments bearing the

oxindole motif and have considerable economical importance. As a

consequence, synthetic methodologies have been developed for the obtention

of these pigments and analogues. Indigo and monothioindigo can be obtained

from the reaction of isatin with P4S10 284. Isoindigos have been prepared by

an acid catalyzed reaction of isatin and oxindole derivatives285-286 and from the

reaction of N-methylisatoic anhydride or N-methylisatin with sodium

phosphonates287-288.

Isoindigos and thioisoindigos have also been prepared from the

reaction of isatin with Lawesson’s reagent289. Indirubins, which are described

as effective antileukemic agents, can be prepared from isatin and indican, a

compound extracted in high yield from Baphicacanthus cusia290, or from isatin

and N-methyl-O-acetylindoxyl 291 and from isatin and 3-hydroxyindole292

Pyrrolo-indigo compounds can be prepared by the condensation of

isatin with pyrrolin-4-ones293; and thionapthene indigo dyes (Thioindigo

Scarlet) are obtained from hydroxythionapthenes and isatin in acidic media294.

In a reverse sense, isatin has been identified as one of the products of the

oxidation of indigo by nitric acid and light. This process may be involved in the

fading of indigo in museum collection objects295 and denim jeans296-297. The

same conversion can be realized by ozonolysis298, acidic bromate299 or by a

chemiluminescent autoxidation of indigo300. N-Methylisatin is also obtained in

the photooxidation of N-methylindole-3-acetic acid301.

Isoindigo, obtained from isatin (74) and oxindole (81), is converted

diastereoselectively into diazacrisenodiones (82) by reduction with Zn/AcOH,

and subsequent acid-catalyzed rearrangement285 as given below.

Page 181: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

177

NH

O

O

(74)

NH

O+CH3COOH, HCl

Heat(100%)

NH

O

HN

O

Zn, HCl

NH

O

HN

OHCl (4mol/ L)

(85 %)

HN

NH

O

OH

H

(81)

(82) An efficient and versatile method for stereoselective synthesis of (E)-

3,3-(diarylmethylene)indolinones (83) by a palladium-catalyzed tandem Heck-

carbocyclization/Suzuki-coupling sequence is presented here as a synthetic

approach towards indolinones302.

NH

O

IR

Pd(PPh3)4, CuTCArB(OH)2, rt

RNH

O

Ar

(83)

A new synthesis of 3-[(4-amido)pyrrol-2-yl]-2-indolinones has been

developed, where the amide side chain was installed prior to pyrrole

formation. This strategy precludes the need to use any coupling reagents to

install the amide side chain. This process includes a zinc-free alternative to

the Knorr pyrrole synthesis303.

5.10 Plan of work

Observed through the literature review, 1,2,4-triazoles and 1,3,4-

thiadiazoles are important from medicinal, industrial and agricultural point of

view. The presence of a mercapto group at 3-position of 1,2,4-triazoles has

been found to show an increase in the antimicrobial activities. Moreover, we

Page 182: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

178

know that a chiral drug interacts with a chiral receptor site; the two

enantiomers of the drug interact differently and may lead to different effects.

In view of all these facts, it was planed to utilize carboxylic acid hydrazides in

the synthesis of substituted 1,2,4-triazoles-3-thione and substituted 1,3,4-

thiadiazoles.

In order to achieve the objectives of the present work, following

synthetic plan was devised (Scheme 5.1).

R OH

O CH3OH

R O

O H2NNH2

H2OR N

H

ONH2

NC S

R'

R

O

NH

HN

S

HN

R'

NaOH

Conc H2SO4

NN

N

NN

S

R SH

NHR

R'

R'

Acid Ester HydrazideIsothiocyanat

Thiosemicarbazide1,2,4-Triazole

1,3,4-Thiadiazole

NH

O

OR''

NH

O

NR''

HN

R

O

5-Haloisatin

Isatin derivatives(Indolinones)

Scheme 5.1

The plan which is illustrated by scheme was based on the following known

principles:

1. Carboxylic acids undergo esterification in acidic medium in the

presence of an alcohol.

2. Esters of carboxylic acids are converted to their respective hydrazides

with hydrazine monohydrate.

Page 183: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

179

3. Isothiocyanates are formed from amines involving salts of

dithiocarbamate as an intermediate.

4. Condensation of carboxylic acid hydrazides with isothiocyanates yields

thiosemicarbazides.

5. Intramolecular dehydrative cyclization of substituted

thiosemicarbazides in basic medium affords the corresponding

substituted 3-mercapto-1,2,4-triazoles.

6. Cyclization of substituted thiosemicarbazides in acidic medium gives

the corresponding substituted 1,3,4-thiadiazoles.

The synthesized hydrazides are also used for the synthesis of oxindols

(2-indolinones), by condensation with halo-isatins in the presence of ethanol

solvent.

The characterization of the synthesized compounds was carried out by

spectroscopic techniques, like UV, IR, 1H NMR, 13C NMR and Mass

spectrometry. The synthesized indolinones, 4-substituted-1,2,4-triazol-3-ones

and thiadiazoles were screened for their antifungal, antibacterial, herbicidal,

insecticidal, fungicidal, plant growth regulatory activity and antiviral activities

Page 184: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

180

Chapter- 6 RESULTS AND DISCUSSION

This chapter deals with the results and discussions of the synthetic aspects of

the target compounds. Firstly the synthesis of carboxylic acid hydrazides was

carried out by the condensation of carboxylic esters of corresponding acids with

hydrazine hydrate (80%). The carboxylic acid hydrazides were then reacted with

2-methoxyphenylisothiocyanate, 4-methoxyphenylisothiocyanate and cyclohexyl-

isothiocyanate to obtain corresponding 1,4-disubstituted thiosemicarbazides.

Each of the isothiocyanate was prepared by the reaction of corresponding amine

with carbon disulphide in the presence of ammonium hydroxide and lead nitrate.

These thiosemicarbazides were subjected to dehydrative cyclization in basic

medium and acidic medium to furnish corresponding 1,2,4-triazoles and 1,3,4-

thiadiazoles.

The synthesized hydrazides were also used for the synthesis of oxindols

(2-indolinones), by condensation with halo-isatins in the presence of ethanol

solvent.

6.1 Synthesis of methyl / ethyl esters 85(a-r)

The methyl esters were synthesized by the reaction of corresponding

substituted benzoic acids with methanol/ethanol in the presence of a catalytic

amount of sulfuric acid 304 (Scheme 6.1). The esters were characterized on the

basis of their physical constants and IR spectral data (Table 6.1).

CH3OH / H+

ORC2H5OH / H+

Reflux 5-6 hrs84(a-r) 85(a-r)

X C-OHO

R

X C-OMeO

R

[Yield = 77- 92%]

Page 185: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

181

a. X = 0 R = 2,5-F2

b. X = 0 R = 3,5-F2

c. X = 0 R = 2,6-F2

d. X = 0 R = 3,4-(OCH3)2

e. X = 0 R = 3,5-(OCH3)2

f. X = 0 R = 2,4-(OCH3)2

g. X = 0 R = 2,6-(OCH3)2

h. X = CH2 R = 4-Cl

i. X = CH2 R = 4-F

j. X = (CH2)2 R = 3,4,5-(OCH3)3

k. X = (CH2)2 R = 4-OCH3

l. X = OCH2 R = 4-Br

m. X = O(CH2)2 R = 2,4-Cl2

n. X = O(CH2)3 R = 2,4-Cl2

o. X = OCH2 R = 2,4-Cl2

p. X = CH2= CH2 R = 4-OCH3

q. X = CH2 R = 3-OCH3

r. X = SCH2 R = 2,4-Cl2

Scheme 6.1: Synthesis of esters 85 (a-r)

Esters 85(a-r) can be characterized by the disappearance of broad peak

of acid in the range of 3200- 2400 cm-1. The IR spectral data of compounds 85(a-r) is tabulated in table 6.1.

Page 186: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

182

Table 6.1: Physical constants and IR spectral data of methyl esters 85(a-r)

νmax (cm-1) Compd M.P (°C) Yield

(%) Rf * Ar–H Sp3 C–H C=O C=C C-O

85a Oil 92 0.72 3060 2942 1726 1585 1290 85b Oil 85 0.50 3058 2940 1727 1582 1309 85c Oil 87 0.71 3061 2943 1728 1590 1313 85d 58-62 79 0.81 3089 2901 1740 1610 1304 85e 41-43 77 0.79 3063 2896 1735 1625 1315 85f 116-121 80 0.74 3085 2890 1738 1596 1325 85g 87-90 81 0.74 3030 2989 1725 1645 1296 85h Oil 78 0.79 3056 2910 1711 1569 1285 85i Oil 83 0.59 3101 3001 1729 1578 1387 85j Oil 89 0.68 3048 3010 1725 1596 1269 85k Oil 79 0.78 3084 2986 1751 1586 1316 85l Oil 89 0.75 3104 2916 1742 1610 1296

85m Oil 82 0.83 3089 2986 1726 1566 1310 85n Oil 83 0.77 3125 2896 1722 1577 1296 85o Oil 82 0.76 3110 2975 1745 1587 1283 85p Oil 78 0.75 3056 2974 1720 1650 1300 85q 125-126 89 0.69 3178 2986 1731 1612 1285 85r Oil 87 0.63 3018 2946 1735 1625 1310 * Petroleum ether: ethylacetate (4:1)

The synthesis of two of the esters 85g and 85q, is also confirmed by x-ray

crystallography, as given below.

6.1.1 Crystal structure of Ethyl 2-(3′-methoxyphenyl)acetate 85q

Ethyl 2-(3′-methoxyphenyl)acetate (85q) (Fig. 6.1) shows no unusual

features. The diherdral angle between the planar ester group and benzene ring is

74.81 (2).

As can be seen from the packing diagram (Fig. 6.2), the Ethyl 2-(3′-

methoxyphenyl)acetate (85q) is stacked along a axis and elongated along the b

axis. Dipole-dipole and Van der waals interactions are effective in the molecular

packing.

Page 187: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

183

Fig. 6.1: Crystal structure of Ethyl 2-(3′-methoxyphenyl)acetate 85q

Fig. 6.2: Packing diagram for Ethyl 2-(3′-methoxyphenyl)acetate

Crystal data C11H14O3 Dx = 1.223 Mg m−3 Mr = 195.23 Melting point: 398(1) K Monoclinic, P21/n Mo Kα radiation λ = 0.71073 Å a = 14.868 Å Cell parameters from 1609 reflections b = 4.851 Å θ = 2.9–23.6º

Page 188: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

184

c = 15.428 Å µ = 0.09 mm−1 β = 107.70º T = 293 (2) K V = 1060.1 Å3 Block, colourless Z = 4 0.40 × 0.30 × 0.30 mm F000 = 420

Geometric parameters (°A,°)

Selected bond lengths: O1—C2 1.596 (6) C6—C10 1.401 (12) O2—C4 1.460 (9) C1—C2 1.369 (9)

C1—C8 1.414 (9) O3—C10 1.344 (10) O3—C12 1.409 (11) C4—C11 1.480 (12)

Selected bond angles:

C10—C7—H7 119.7 C2—C1—C8 133.9 (6) C8—C7—H7 119.7

C7—C10—C6 119.7 (7) O2—C4—C11 106.4 (6)

Selected torsional bond angles: C9—C5—H5 119.2 H12A—C12—H12B 109.5 C5—C6—C10 118.7 (7) C10—C7—C8 120.6 (7) C4—O2—C2—C1 176.1 (5) C7—C8—C9—C5 1.2 (12) C4—O2—C2—O1 −5.2 (9)

C6—C5—C9—C8 −1.6 (14) C12—O3—C10—C7 170.4 (8) C9—C5—C6—C10 2.2 (13) C10—C7—C8—C1 178.0 (7) C2—C1—C8—C9 175.6 (7) C5—C6—C10—C7 −2.5 (11) C2—C1—C8—C7 −4.0 (12)

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Murat Tas and Okan Zafer Yesilel; Crystal structure of Ethyl-2-(3-

methoxyphenyl)acetate; Acta Cryst.; 2007, E63 ,o4678.

Page 189: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

185

6.1.2 Crystal structure of Methyl 2,6-dimethoxybenzoate (85g)

A perspective view of methyl 2,6-dimethoxybenzoate (85g) is shown in

Fig. 6.3. Bond lengths and angles can be regarded as normal. The ester group

is perpendicular by 91.88 (19) out of the plane of the central aromatic ring.

Fig. 6.3: Crystal structure of Methyl 2,6-dimethoxybenzoate (85g)

Fig. 6.4: Packing diagram for Methyl 2,6-dimethoxy benzoate

Page 190: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

186

π-π Stacking interaction can be observed in the crystal packing (Fig. 6.4).

Carboxylate atom C7 is located above the centre of the aromatic ring of a

neighboring parallel molecule (at x −1, y, z) at a distance of 3.585 A °.

Crystal data C10H12O4 Dx = 1.317 Mg m−3 Mr = 196.20 Melting point: 450(1) K Orthorhombic, Pbca Mo Kα radiation λ = 0.71073 Å a = 7.2306 (9) Å Cell parameters from 1609 reflections b = 14.1058 (17) Å θ = 2.9–23.6º c = 19.403 (2) Å µ = 0.10 mm−1 V = 1978.9 (4) Å3 T = 100 (2) K Z = 5 Block, colourless F000 = 832 0.40 × 0.30 × 0.30 mm

Geometric parameters (°A, °)

Selected Bond lengths: O1—C7 1.3335 (16) C4—C5 1.379 (2) O1—C8 1.4548 (17) O4—C6 1.3680 (17)

O4—C10 1.4397 (17) C1—C6 1.3951 (19) C1—C2 1.398 (2) C1—C7 1.498 (2)

Selected Bond angles: C7—O1—C8 116.06 (12) O2—C7—O1 123.88 (14) C2—O3—C9 117.32 (12) O2—C7—C1 124.86 (13)

H8B—C8—H8C 109.5 C4—C3—C2 118.84 (16) C5—C4—C3 122.38 (16) C4—C5—C6 118.39 (16)

Selected torsional bond angles: C9—O3—C2—C3 −1.7 (2) C4—C5—C6—O4 179.56 (13) C6—C1—C2—O3 −179.47 (12) C2—C1—C6—O4 179.59 (13)

C7—C1—C2—C3 178.67 (13) C7—C1—C6—C5 −177.57 (13) O3—C2—C3—C4 179.34 (13) C8—O1—C7—O2 −2.0 (2)

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Mohammad Azad Malik and Madeleine Helliwell; Crystal structure of

Methyl 2,6-dimethoxybenzoate; Acta Cryst. 2007, E63, o3027.

Page 191: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

187

6.2 Synthesis of hydrazides 86(a-r)

Methyl / Ethyl esters 85(a-r) of substituted benzoic acids were converted

to the corresponding hydrazides 86(a-r) by refluxing with hydrazine hydrate

(80%) in methanol305 (Scheme 3.2). The synthesized hydrazides 86(a-r) were

recrystallized from aqueous ethanol and characterized by their physical

constants, IR (Table 6.2), 1H NMR, 13C NMR (Table 6.3), Mass spectrometry

and X-ray crystallography.

[Yield = 72 - 88%]85(a-r) 86(a-r)X C-OMe

OR

X C-NHO

R

NH2

NH2-NH2.H2OCH3OHReflux4-7hrs

Scheme 6.2: Synthesis of substituted hydrazides 86 (a-r)

a. X = 0 R = 2,5-F2

b. X = 0 R = 3,5-F2

c. X = 0 R = 2,6-F2

d. X = 0 R = 3,4-(OCH3)2

e. X = 0 R = 3,5-(OCH3)2

f. X = 0 R = 2,4-(OCH3)2

g. X = 0 R = 2,6-(OCH3)2

h. X = CH2 R = 4-Cl

i. X = CH2 R = 4-F

j. X = (CH2)2 R = 3,4,5(OCH3)3

k. X = (CH2)2 R = 4-OCH3

l. X = OCH2 R = 4-Br

m. X = O(CH2)2 R = 2,4-Cl2

n. X = O(CH2)3 R = 2,4-Cl2

o. X = OCH2 R = 2,4-Cl2

p. X = CH2= CH2 R = 4-OCH3

q. X = CH2 R = 3-OCH3

r. X = SCH2 R = 2,4-Cl2

The IR spectra of hydrazides 86(a-r) exhibited a characteristic absorption

band for primary NH2, in the region 3547-3309 cm-1, while absorption band for

the secondary NH was observed in the range 3476-3198 cm-1. A strong

absorption in the region 1662-1618 cm-1 was assigned to the carbonyl group of

amide linkage. The IR spectral data of hydrazides 86(a-r) is given in table 6.2.

Page 192: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

188

Table 6.2: Physical constants and IR spectral data of hydrazides 86(a-r)

υmax (cm-1) S.No m.p (°C) Yield (%) Rf* NH2 NH C=O C=C C–O

86a 141-142 78 0.80 3312 3312 b 1626 1597 1249 86b 158-159 74 0.87 3326 3286 1628 1595 1262 86c 244-246 88 0.75 3316 3316 b 1632 1579 1260 86d 118 82 0.81 3335 3296 1661 1596 1255 86e 165-166 73 0.78 3410 3325 1656 1578 1268 86f 101-102 72 0.80 3350 3210 1668 1560 1256 86g 160-161 79 0.87 3361 3256 1659 1589 1254 86h 102-104 79 0.75 3396 3241 1654 1596 1256 86i 158-160 81 0.81 3385 3274 1674 1574 1253 86j 121-122 78 0.78 3345 3196 1648 1585 1241 86k 125-126 78 0.80 3341 3341 1659 1574 1248 86l 110-112 74 0.87 3252 3252b 1658 1572 1249

86m 164-165 82 0.75 3345 3210 1654 1574 1254 86n 154-155 78 0.81 3385 3202 1652 1547 1247 86o 63-65 74 0.78 3374 3216 1663 1569 1257 86p 151-152 88 0.80 3341 3189 1639 1547 1251 86q 204-206 82 0.87 3298 3195 1658 1601 1239 86r 60-62 72 0.75 3347 3347b 1645 1540 1281

*petroleum ether: ethylacetate (4:1) b broad band

The formation of hydrazide 86(a-r) from the respective methyl/ethyl esters

was confirmed in the 1H NMR spectra by the appearance of NH2 proton signal in

the region of 2.00-2.50 ppm and NH proton signal in the region of 8.50- 9.98ppm

and by the disappearance of –OCH3 proton signals in the region of 3.77-

3.90ppm. In all these compounds 86(a-r), the integration curve indicated three

protons in the aromatic region confirming the presence of phenyl ring. In all these

compounds having two fluorine atoms as a substituent, splitting due to coupling

between fluorine and the hydrogens was also observed. The structures are futher

confirmed by 13C NMR (Table 6.3), mass spectrometry and X-ray

crystallography. The detail of spectral data of more hydrazides is given in chapter

7 (Experimental).

Page 193: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

189

Table 6.3: 1H NMR and 13C NMR spectral data of 3,5-Difluorophenylhydrazides

O

HN

NH2

12 3

4

56

F

F

δ (ppm) and multiplicity Carbon

1H NMR 13C NMR

C=O ---- 164.3 OCH3 ---- 56.3 C-1 ---- 137.18 C-2 7.39-7.55(m) 107.02 C-3 ---- 163.58 C-4 7.39-7.55(m) 110.74 C-5 ---- 161.02 C-6 7.39-7.55(m) 110.57 NH 9.98 (s) ---- NH2 4.60(s) ----

6.2.1 Crystal structure of 3,5-difluorobenzohydrazide (86b)

The molecular structure of 3,5-difluorobenzohydrazide (86b) is shown in

Fig. 6.5. Bond distances and angles are within expected ranges. The hydrazidic

group C7/O1/N1/N2 is planar (maximum displacement being 0.007 (3) Å for C7)

and nearly coplanar with the benzene ring [the dihedral angle being 9.27 (10) °].

The crystal packing is stabilized by N—H···O and N—H···N intermolecular

hydrogen interactions (Fig. 6.6).

Page 194: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

190

Fig. 6.5: Crystal structure of 3,5-Difluorobenzohydrazide (86b)

Fig. 6.6: Packing diagram showing the hydrogen bonding.

Crystal data C7H6F2N2O F000 = 352 Mr = 172.14 Dx = 1.558 Mg m−3 Orthorhombic, P212121 Melting point: 374(1) K Hall symbol: P 2ac 2ab Mo Kα radiation λ = 0.71073 Å a = 3.8635 (4) Å µ = 0.14 mm−1 b = 6.4199 (6) Å T = 293 (2) K c = 29.589 (3) Å Block, pale yellow V = 733.89 (13) Å3 0.42 × 0.30 × 0.26 mm Z = 4

Page 195: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

191

Geometric parameters (°A, °)

Selected bond lengths: C1—C2 1.380 (3) C5—C6 1.385 (2) C6—C7 1.500 (2) C7—N1 1.337 (2)

N1—H1 0.88 (2) C4—F2 1.355 (2) C4—C5 1.382 (2) N2—H3 0.85 (2)

Selected bond angles: C2—C1—C6 118.14 (16) C5—C6—C1 120.34 (14) C5—C6—C7 116.48 (14) F1—C2—C3 118.51 (17)

N2—N1—H1 114.8 (12) F2—C4—C5 117.72 (19) N1—N2—H2 107.8 (14) C3—C4—C5 123.62 (17)

Selected torsional bond angles: C6—C1—C2—F1 178.82 (17) C4—C5—C6—C7 −177.92 (18) F1—C2—C3—C4 −178.66 (18) C1—C6—C7—O1 −178.34 (16)

C2—C3—C4—C5 −0.1 (3) C5—C6—C7—N1 178.82 (15) F2—C4—C5—C6 178.36 (19) C6—C7—N1—N2 −173.47 (14)

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N1—H1···N2i 0.88 (2) 2.14 (2) 2.9733 (19) 158.4 (16) N2—H2···O1ii 0.93 (2) 2.05 (2) 2.9737 (19) 175 (2) N2—H3···O1 0.85 (2) 2.38 (2) 2.7240 (17) 104.6 (17) N2—H3···O1iii 0.85 (2) 2.42 (2) 3.024 (2) 128.8 (17)

Symmetry codes: (i) x−1/2, −y+3/2, −z; (ii) x−1/2, −y+5/2, −z

(iii) x+1/2, −y+5/2, −z

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Muhammad Zareef and Wai-Yeung Wong; Crystal structure of 3,5-

Difluorobenzohydrazide , Acta Cryst, 2007, E63, o1176–o1177.

Page 196: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

192

6.2.2 Crystal structure of 2,6-dimethoxybenzohydrazide (86g)

Fig. 6.7: Crystal structure of 2,6-Dimethoxybenzohydrazide (86g)

The molecular structure of 2,6-dimethoxybenzohydrazide (86g) is shown

in Fig 6.7. Bond distances and angles are within expected ranges. The

hydrazidic group C9/O3/N1/N2 is planar (maximum displacement being 0.007 (3)

Å for C8) and perpendicular with the benzene ring [the dihedral angle being

88.27 (10) °]. Crystal data C9H12N2O3 Dx = 1.257 Mg m−3 Mr = 196.21 Melting point: 244(2) K Orthorhombic, Pbca Mo Kα radiation λ = 0.71073 Å a = 7.2598 (5) Å Cell parameters from 1520 reflections b = 14.2558 (11) Å θ = 2.7–24.9º This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama and Wen Tong Chen; Crystal structure of 2,6-Dimethoxybenzohydrazide

Acta Cryst. 2007, E63, o2892.

Page 197: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

193

c = 20.0412 (11) Å µ = 0.10 mm−1 V = 2074.1 (2) Å3 T = 293 (2) K Z = 8 Block, colorless F000 = 832 0.16 × 0.14 × 0.06 mm

Geometric parameters (°A, °)

Selected bond lengths: O1—C2 1.366 (3) C2—C3 1.393 (4) C7—H7B 0.9600 N2—H2A 0.8600 C7—H7C 0.9600

C8—H8A 0.9600 C1—C2 1.393 (3) C8—H8B 0.9600 C1—C9 1.506 (3) C8—H8C 0.9600

Selected bond angles: C2—O1—C7 118.4 (3) C4—C5—H5A 120.7 C9—N2—N1 118.5 (2) H7B—C7—H7C 109.5 O1—C2—C3 125.5 (3) O2—C8—H8B 109.5 C3—C2—C1 119.5 (3)

H8A—C8—H8B 109.5 C2—C3—C4 118.5 (3) O2—C8—H8C 109.5 C2—C3—H3A 120.7 C3—C4—H4A 118.6 N2—C9—C1 112.7 (2) C4—C5—C6 118.6 (3)

6.2.3 Crystal structure of 3,4-dimethoxybenzohydrazide (86d)

Fig. 6.8: Crystal structure of 3,4-Dimethoxybenzohydrazide (86d)

Page 198: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

194

The molecular structure of (86d) is shown in Fig. 6.8. Bond distances and

angles are within expected ranges. The dihedral angle between the planar

hydrazidic group (C7/O1/N1/N2) and the benzene ring (C1—C6) is 63.27 (3) °. The crystal structure is stabilized by N–H···O hydrogen bonding (Fig. 6.9).

Fig. 6.9: Packing diagram showing the hydrogen bonding

Crystal data C9H12N2O3 Dx = 1.396 Mg m−3

Dm = 1.375 Mg m−3 Mr = 196.21 Melting point: 391(1) K Monoclinic, P2(1)/c Mo Kα radiation λ = 0.71073 Å a = 13.610 (3) Å Cell parameters from 927 reflections b = 8.9130 (19) Å θ = 2.8–25.1º c = 7.9780 (17) Å µ = 0.11 mm−1 β = 105.266 (4) º T = 100 (2) K V = 933.6 (3) Å3 Block, white Z = 4 0.25 × 0.20 × 0.20 mm F000 = 416

Page 199: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

195

Geometric parameters (°A, °)

Selected bond lengths: C1—C6 1.385 (3) C7—N1 1.334 (2) C1—C2 1.402 (2) C8—O2 1.434 (2)

C8—H8B 0.9800 C2—H2 0.9500 C3—C4 1.413 (3) C9—H9A 0.9800

Selected bond angles: C6—C1—C2 119.75 (18) O2—C8—H8A 109.5 C6—C1—C7 122.13 (18) O2—C8—H8B 109.5 C4—C5—C6 120.17 (18)

C7—N1—N2 121.93 (17) C1—C6—C5 120.27 (19) N1—N2—H2A 105.5 (14) C1—C6—H6 119.9 N1—N2—H2B 105.3 (14)

Selected torsional bond angles: C6—C1—C2—C3 1.8 (3) C4—C5—C6—C1 −0.9 (3) C7—C1—C2—C3 177.49 (17) C6—C1—C7—O1 147.43 (18) C1—C2—C3—O2 178.76 (16) C2—C1—C7—O1 −28.1 (3)

O2—C3—C4—C5 179.36 (16) C1—C7—N1—N2 175.03 (17) C2—C3—C4—C5 −1.0 (3) C2—C3—O2—C8 10.0 (3) O3—C4—C5—C6 −179.15 (18) C4—C3—O2—C8 −170.31 (15)

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A

N1—H1···O1i 0.88 2.10 2.894 (2) 150

N2—H2A···O1ii 0.90 (2) 2.17 (2) 2.944 (2) 144.0 (18)

Symmetry codes: (i) −x, y+1/2, −z−1/2; (ii) x, −y+3/2, z−1/2.

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Mohammad Azad Malik and James Raftery; Crystal structure of 3,4-

Dimethoxybenzohydrazide; Acta Cryst. 2007, E63, o3026.

Page 200: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

196

6.2.4 Crystal structure of 2-(2′,4′-dichlorophenylsulfanyl)acto-

hydrazide (86r)

Fig. 6.10: Crystal structure of 2-(2′,4′-Dichlorophenylsulfanyl)actohydrazide (86r)

The molecular structure of (86r) is shown in Fig. 6.10. Bond distances and

angles are within expected ranges. The dihedral angle between the planar

hydrazidic group (C8/O1/N1/N2) and benzene ring (C1—C6) is 91.07 (3) °. Two

centrosymmetrically related N1—H1A···O1 (N1···O1, 3.078 A, H1A···O1, 2.666 A,

N1—H1A···O1, 110.8) hydrogen bonds form a dimer (Fig. 6.11).

Fig. 6.11: Packing diagram showing the hydrogen bonding

Page 201: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

197

Crystal data

2(C8H8Cl2N2O1S1) Z = 2 Mr = 251.13 F000 = 240 Triclinic, P1 Dx = 1.657 Mg m−3 Hall symbol: -P 1 Melting point: 333(2) K a = 7.350 (5) Å Mo Kα radiation λ = 0.71073 Å b = 8.133 (6) Å Cell parameters from 1520 reflections c = 8.545 (6) Å θ = 2.7–24.9º α = 94.802 (10) º µ = 0.82 mm−1 β = 90.140 (9) º T = 293 (2) K γ = 98.492 (10) º Block, colourless V = 503.4 (6) Å3 0.15 × 0.14 × 0.14 mm

Geometric parameters (°A, °)

Selected bond lengths: Cl1—C6 1.749 (3) C1—C2 1.376 (4) Cl2—C4 1.729 (3) C1—H1C 0.9300

N1—H1B 0.8600 C5—H5A 0.9300 N2—C8 1.334 (4) C7—C8 1.502 (4)

Selected bond angles: C3—S1—C7 117.5 (2) C5—C4—Cl2 119.6 (2) N2—N1—H1A 120.0 S1—C7—C8 110.3 (2) C2—C1—H1C 120.3 S1—C7—H7A 109.6 C1—C2—C3 121.7 (3)

C8—C7—H7A 109.6 C1—C2—H2B 119.2 S1—C7—H7B 109.6 C3—C2—H2B 119.2 C8—C7—H7B 109.6 S1—C3—C2 126.5 (3) H7A—C7—H7B 108.1

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama and Wen-Tong Chen Crystal structure of 2-(2,4-Dichlorophenylsulfanyl)-

acetohydrazide ; Acta Cryst. 2007, E63, o2932.

Page 202: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

198

6.2.5 Crystal structure of (E)-3-(4′-methoxyphenyl)acrylo-

hydrazide (86p)

Fig. 6.12: Crystal structure of (E)-3-(4′-Methoxyphenyl)acrylohydrazide (86p)

The molecular structure of (E)-3-(4′-methoxyphenyl)acrylohydrazide (86p)

is shown in Fig. 6.12. Bond distances and angles are within expected ranges.

The dihedral angle between the planar hydrazidic group (C10/O2/N1/N2) and the

benzene ring (C1—C6) is 73.93 (3) °.

Crystal data

C10H12N2O2 Dx = 1.213 Mg m−3 Dm = 1.213 Mg m−3 Dm measured by not measured

Mr = 192.20 Melting point: 477(2) K Monoclinic, P2 (1)/c Mo Kα radiation λ = 0.71069 Å a = 18.661 (5) Å Cell parameters from 691 reflections b = 4.842 (5) Å θ = 3.5–23.6º c = 12.041 (5) Å µ = 0.09 mm−1 β = 106.774 (5) º T = 293 (2) K V = 1041.7 (12) Å3 Block, colorless Z = 4 0.30 × 0.20 × 0.10 mm F000 = 400

Page 203: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

199

Geometric parameters (°A, °)

Selected bond lengths: O2—C10 1.233 (2) C4—C3 1.377 (3) N1—C10 1.3319 (19) C4—C8 1.512 (2)

N1—N2 1.4132 (17) C8—H8 0.9700 O1—C7 1.424 (3) C7—H7A 0.9600

Selected bond angles: C10—N1—N2 123.06 (12) C4—C5—H5 118.9 C10—N1—H1N 117.4 (12) O2—C10—N1 121.96 (14) C1—C6—H6 120.0 C6—C1—O1 124.95 (17)

C4—C8—H8 108.9 C6—C1—C2 119.31 (16) O1—C7—H7A 109.5 O1—C1—C2 115.74 (16) O1—C7—H7B 109.5 C5—C4—C3 116.80 (17)

Selected torsional bond angles: N2—N1—C10—O2 −1.4 (2) C2—C1—C6—C5 −0.5 (3) O2—C10—C9—C8 40.0 (2)

C1—C2—C3—C4 −0.4 (3) C3—C4—C8—C9 82.5 (3) O1—C1—C2—C3 −178.76 (18)

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Zhong-Min Su; Crystal structure of (E)-3-(4′-Methoxyphenyl)acrylo-

hydrazide; Acta Cryst. 2007, E63, o2989.

Page 204: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

200

6.2.6 Crystal structure of 3-(3′,4′,5′-trimethoxyphenyl)propane-

hydrazide (86j)

Fig. 6.13: Crystal structure of 3-(3′,4′,5′-Trimethoxyphenyl)propanohydrazide

(86j)

The molecular structure of 3-(3′,4′,5′-trimethoxyphenyl)propanohydrazide

(86j) is shown in Fig. 6.13. Bond distances and angles are within expected

ranges. The dihedral angle between the planar hydrazidic group (C9/O1/N1/N2)

and the benzene ring (C1—C6) is 61.23 (2) °.

X-ray analysis reveals that the asymmetric unit contains two independent

molecules. N—H···O and C—H···O hydrogen bonds link the molecules into

Page 205: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

201

layers. Molecules in adjacent layers are linked via N—H···O hydrogen bonds

(Fig. 6.14).

Fig. 6.14: Packing diagram showing the H-bonding Crystal data C12H18N2O4 Dx = 1.364 Mg m−3

Dm = 1.323 Mg m−3 Dm measured by not measured

Mr = 254.28 Melting point: 398(1) K Monoclinic, P2 (1)/n Mo Kα radiation λ = 0.71073 Å a = 9.7770 (14) Å Cell parameters from 2527 reflections b = 20.189 (3) Å θ = 2.4–24.6º c = 12.7300 (19) Å µ = 0.10 mm−1 β = 99.618 (3) º T = 100 (2) K V = 2477.4 (6) Å3 Plate, white Z = 8 0.20 × 0.20 × 0.05 mm F000 = 1088

Geometric parameters (°A, °)

Selected bond lengths: C1—C6 1.387 (2) C14—H14 0.9500

C1—C2 1.389 (2) C16—O7 1.3814 (19)

Page 206: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

202

C4—O3 1.3844 (18) C18—H18 0.9500

C4—C5 1.392 (2) C19—C20 1.529 (2)

Selected bond angles: C6—C1—C2 119.66 (16) O7—C16—C15 120.21 (16) C6—C1—C7 123.41 (16) O7—C16—C17 120.41 (16) C20—C19—H19A 108.3 O4—C5—C4 115.25 (15)

O1—C9—N1 122.40 (16) O7—C23—H23A 109.5 O1—C9—C8 122.22 (16) O7—C23—H23B 109.5 N1—C9—C8 115.37 (16)

Selected torsional bond angles: C6—C1—C2—C3 −1.8 (3) C7—C1—C2—C3 174.91 (16) C1—C2—C3—O2 −178.38 (16) C1—C2—C3—C4 0.0 (3) O2—C3—C4—O3 −2.5 (2) C2—C3—C4—O3 179.08 (14) O3—C4—C5—C6 −179.06 (15) C3—C4—C5—C6 −1.9 (2)

O5—C21—N3—N4 −1.5 (3) C16—C15—O6—C22 169.91 (15) C13—C14—C15—C16 0.8 (3) C14—C15—O6—C22 −9.7 (2) O6—C15—C16—O7 −3.3 (2) C17—C16—O7—C23 −85.0 (2

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N4—H4B···O6i 0.924 (18) 2.491 (19) 3.401 (2) 168.2 (15) N2—H2B···O5 1.016 (19) 2.110 (19) 3.045 (2) 152.1 (14) N2—H2A···O3ii 0.883 (17) 2.618 (17) 3.453 (2) 158.2 (15) N2—H2A···O2ii 0.883 (17) 2.377 (17) 3.067 (2) 135.2 (14) N4—H4A···O1 0.997 (19) 2.131 (19) 3.084 (2) 159.5 (15) N3—H3···O1iii 0.88 2.02 2.8914 (18) 171 N1—H1···O5iv 0.88 2.01 2.8719 (18) 166 Symmetry codes: (i) −x+3/2, y−1/2, −z+1/2; (ii) −x+3/2, y−1/2, −z+3/2; (iii) −x+2,

−y, −z+1; (iv) −x+1, −y, −z+1.

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Mohammad Azad Malik and James Raftery; Crystal structure of 3-

(3′,4′,5′-Trimethoxyphenyl)propanohydrazide; Acta Cryst., 2007, E63 , o3025.

Page 207: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

203

6.2.7 Crystal structure of 3-(4′-methoxyphenyl)propanohydrazide

(86k)

Fig. 6.15: Crystal structure of 3-(4′-Methoxyphenyl)propanohydrazide (86k)

The molecular structure of 3-(4′-methoxyphenyl)propanohydrazide (86k)is

shown in Fig. 6.15. Bond distances and angles are within expected ranges. The

dihedral angle between the planar hydrazidic group (C9/O1/N1/N2) and the

benzene ring (C1—C6) is 81.27 (3) °. The crystal structure is stabilized by N–

H···O and N–H···N hydrogen bonding (Fig. 6.16).

Fig. 6.16: Packing diagram showing the H-bonding

Page 208: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

204

Crystal data C10H14N2O2 F000 = 416 Mr = 194.23 Dx = 1.276 Mg m−3

Dm = 1.253 Mg m−3 Dm measured by not measured

Monoclinic, P21/c Melting point: 383(2) K Hall symbol: -P 2ybc Mo Kα radiation λ = 0.71073 Å a = 18.519 (9) Å Cell parameters from 691 reflections b = 4.816 (2) Å θ = 3.4–23.6º c = 11.884 (6) Å µ = 0.09 mm−1 β = 107.521 (7) º T = 100 (2) K V = 1010.7 (8) Å3 Block, colorless Z = 4 0.30 × 0.20 × 0.10 mm

Geometric parameters (°A, °) Selected bond lengths: C1—C2 1.390 (5) C7—H7B 0.9900 C1—C6 1.393 (4) C8—C9 1.498 (5)

C1—C7 1.502 (5) C10—H10B 0.9800 C5—C6 1.381 (4) C10—H10C 0.9800

Selected bond angles: C2—C1—C6 116.4 (3) H7A—C7—H7B 107.8 C2—C1—C7 121.9 (3) O2—C4—C5 115.2 (3) O2—C10—H10B 109.5

C5—C6—C1 122.3 (3) C5—C6—H6 118.8 C9—N1—N2 122.9 (3) C1—C6—H6 118.8 C4—O2—C10 116.5 (3)

Selected torsional bond angles: C6—C1—C2—C3 1.2 (5) C2—C1—C7—C8 −98.0 (4) C7—C1—C2—C3 −178.8 (3) C6—C1—C7—C8 82.1 (4)

C3—C4—O2—C10 −4.6 (5) C2—C1—C6—C5 −1.0 (5) C5—C4—O2—C10 174.4 (3) C7—C1—C6—C5 179.0 (3)

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Mohommad Azad Malik and James Raftery; Crystal structure of 3-(4′-

Methoxyphenyl)propanohydrazide; Acta Cryst. 2007, E63 , o3061.

Page 209: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

205

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N1—H1···O1i 0.88 2.04 2.883 (3) 159 N2—H2A···N2ii 0.89 (3) 2.35 (4) 3.174 (4) 154 (3) N2—H2B···O1iii 0.88 (3) 2.28 (4) 3.112 (4) 158 (3)

Symmetry codes: (i) x, y+1, z; (ii) −x+1, y−1/2, −z+3/2; (iii) −x+1, −y+1, −z+1.

6.3 Synthesis of isothiocyanate 87(a-c)306

Three substituted phenyl isothiocyanates namely 2-methoxyphenyl-

isothiocyanate (87a), 4-methoxyphenylisothiocyanate (87b) and cyclohexyl-

isothiocyanate (87c) were prepared by the reaction of corresponding amine with

ammonical carbon disulphide followed by the reaction with lead nitrate. The

isothiocyanates were finally recovered by steam distillation. The general

synthetic scheme is given below.

[Yield = 47 - 70%]

+ CS2 MeOHNH4OHRHN

S

S-NH4+

R NC

S

Stirring12hr

Pb(NO3)2Overnight stirring

+ +

Isothiocyanate

RNH2

87a) R = 2-MeO(C6H4)-87b) R = 4-MeO(C6H4)-87c) R = -C6H11

Amine

87(a-c)

Scheme 6.3: Synthesis of Isothiocyanate 87 (a-c)

All the three isothiocyanates 87(a-c) were obtained in about 50% yield. All

isolated isothiocyanates were purified by column chromatography on silica gel

column in acetone: hexane (7:3). Each isolated isothiocyanate showed a single

spot on TLC which indicated its purity. All the three isothiocyanates showed

Page 210: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

206

characteristic IR absorption band in the region of 2106-2035 cm-1. Thus presence

of N=C=S group in the IR spectrum was indicated by a broad intense peak at

2106 cm-1.

Table 6.4: IR data of Cyclohexyl- and substituted methoxyphenylisothiocyanate

87(a-c)

The formation of isothiocyanates 87(a-c) from the amine were confirmed by 1H NMR spectra by the absence of any proton signal for N-H of the amine. In

case of substituted methoxyphenylisothiocyanate, a single peak for methoxy

protons appeared at 3.79 ppm. The 1H NMR spectral data of isothiocyanate

87(a-c) are given in table 6.5. The 13C NMR spectra of isothiocyanates show

absorption at 155-165ppm due to carbon of isothiocyanate group. In case of

substituted methoxyisothiocyanates a peak in range of 45-55ppm is observed.

The 13C NMR data of isothiocyanates 103(a-e) is also given in table 6.5.

The structure is also confirmed by mass spectrometry. These Isothiocyanates

87(a-c) give M+2 and M+4 peaks due to presence of sulphur atom. The mass

spectral data from isotopic peaks confirmed the formation of Isothiocyanate. In

case of isothiocyanates molecular ion peak is base peak. The molecular ion peak

for the methoxyphenylisothiocyanate is 165 (100%) and 141(100%) for

cyclohexyl- isothiocyanate.

υmax (cm-1) Code M.P.

(°C) Yield Rf sp3 C-H stretching

sp2 C-H stretching N=C=S C=C

87a oil 49% 0.79 2967 & 2940 3069 2035

broad 1591, 1495 &

1457

87b oil 47% 0.81 2957,

2907 & 2836

3053 2106 broad

1603, 1503 & 1460

87c oil 50% 0.82 2941 3063 2065 broad ----

Page 211: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

207

Table 6.5: 1H NMR data of Cyclohexyl- and substituted methoxyphenyl -

isothiocyanate 87 (a-c)

1H NMR

δ (ppm)

13C NMR

δ (ppm) Code Substitutent

-OCH3

protons

Aromatic

protons

Aliphatic

carbon

Aromatic

carbon

Isothio-

cyanate

carbon

87a 2-methoxyphenyl 3.29 6.89-7.28 51.5 116.50-135.54 162.51

87b 4-methoxyphenyl 3.24 6.81- 7.28 55.58 114.33-133.80 158.57

87C Cyclohexyl ---- 1.32-1.88 ---- 23.23-33.20 129.58

6.4 Synthesis of thiosemicarbazides 88(a-w)

The thiosemicarbazides 88(a-w) were synthesized by the reaction of the

corresponding carboxylic acid hydrazides and isothiocyanate in methanol307-308

(Scheme 6.3). Eighteen different hydrazides 86(a-r) were selected to be

condensed with three substituted isothiocyanate to bring diversity in biological

activity of the final products. The thiosemicarbazides are intermediate for the

synthesis of heterocycles (1,2,4-triazoles and 1,3,4-thiadiazoles).

[Yield = 78 - 85%]86(a-r) 87(a-c) 88(a-w)

X C-NHO

R

NH2 +R′

NC S

O

NH

HN

S

HN

R′Reflux

6 hrsX

R

Scheme 6.4: Synthesis of substituted Thiosemicarbazides 88 (a-w)

88a. X = 0 R = 3,5-F2 R′ = -C6H11 88b. X = O(CH2)2 R = 2,4-Cl2 R′ = -C6H11 88c. X = 0 R = 3,4-(OCH3)2 R′ = -C6H11 88d. X = OCH2 R = 2,4-Cl2 R′ = -C6H11

Page 212: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

208

88e. X = (CH2)2 R = 3,4,5-(OCH3)3 R′ = -C6H11 88f. X = (CH2)2 R = 4-OCH3 R′ = -C6H11 88g. X = OCH2 R = 4-Br R′ = 2-MeO(C6H4)- 88h. X = 0 R = 2,5-F2 R′ = 2-MeO(C6H4)- 88i. X = OCH2 R = 2,4-Cl2 R′ = 2-MeO(C6H4)- 88j. X = 0 R = 3,5-F2 R′ = 2-MeO(C6H4)- 88k. X = 0 R = 3,4-(OCH3)2 R′ = 2-MeO(C6H4)- 88l. X = 0 R = 2,6-(OCH3)2 R′ = 2-MeO(C6H4)- 88m. X = 0 R = 3,5-(OCH3)2 R′ = 2-MeO(C6H4)- 88n. X = (CH2)2 R = 3,4,5(OCH3)3 R′ = 2-MeO(C6H4)- 88o. X = (CH2)2 R = 4-OCH3 R′ = 2-MeO(C6H4)- 88p. X = O(CH2)2 R = 2,4-Cl2 R′ = 4-MeO(C6H4)- 88q. X = O(CH2)3 R = 2,4-Cl2 R′ = 4-MeO(C6H4)- 88r. X = (CH2)2 R = 4-OCH3 R′ = 4-MeO(C6H4)- 88s. X = OCH2 R = 4-Br R′ = 4-MeO(C6H4)- 88t. X = OCH2 R = 2,4-Cl2 R′ = 4-MeO(C6H4)- 88u. X = 0 R = 2,5-F2 R′ = 4-MeO(C6H4)- 88v. X = (CH2)2 R = 3,4,5-(OCH3)3 R′ = 4-MeO(C6H4)- 88w. X = 0 R = 3,4-(OCH3)2 R′ = 4-MeO(C6H4)-

The synthesized compounds were recrystallized from a mixture of ethyl

acetate and petroleum ether. The physical data of compounds 88(a-w) is

tabulated in table 6.6. The conversion to thiosemicarbazides 88(a-w) was

indicated in the IR spectra by the appearance of a carbonyl absorption in the

region of 1692-1665 cm-1 and an absorption in the region 1246-1224 cm-1 for C=S

group. The characteristic absorption bands for three secondary N–H groups were

observed in the region of 3388-3141 cm-1. The IR spectral data of the

synthesized thiosemicarbazides 88(a-w) is presented in table 6.6.

Page 213: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

209

Table 6.6: Physical constants and IR spectral data of Thiosemicarbazides 88(a-w)

υmax (cm-1) Compd mp (°C) *Rf Yield (%) N-H C=O C=S C=C

88a 131-133 0.37 85 3250-3145 1653 1265 1600, 1568

88b 149-150 0.34 83 3271-3149 1659 1251 1612, 1585

88c 185-186 0.38 80 3351-3115 1666 1239 1613, 1561

88d 189-190 0.35 79 3290-3009 1654 1255 1607, 1560

88e 174-176 0.37 84 3400-3145 1658 1244 1598, 1547

88f 133-135 0.34 85 3411-3205 1673 1242 1625, 1588

88g 165-167 0.34 80 3385-3216 1685 1255 1615, 1566

88h 153-154 0.37 78 3285-3210 1670 1245 1605, 1549

88i 156-157 0.40 85 3385-3216 1671 1248 1586, 1539

88j 149-150 0.39 83 3315-3116 1645 1265 1587, 1542

88k 173-175 0.41 80 3410-3016 1660 1251 1587, 1565

88l 180-182 0.35 79 3400-3260 1663 1261 1625, 1541

88m 165-167 0.37 84 3305-3011 1667 1250 1603, 1561

88n 174-175 0.40 85 3445-3211 1656 1255 1621, 1545

88o 189-191 0.34 80 3383-3116 1668 1235 1601, 1520

88p 186-187 0.37 78 3250-3100 1658 1247 1632, 1568

88q 155-156 0.39 85 3311-3019 1670 1257 1604, 1555

88r 165-167 0.35 83 3432-3210 1675 1252 1605, 1546

88s 141-143 0.42 80 3326-3100 1663 1254 1599, 1525

88t 134-135 0.35 79 3142-3001 1675 1248 1605, 1542

88u 119-120 0.37 84 3254-3006 1663 1245 1605, 1509

88v 164-165 0.34 85 3330-3016 1645 1236 1625, 1555

88w 148-149 0.41 80 3400-3180 1668 1262 1596, 1524

*Petroleum ether: Acetone; (6:4)

Formation of these thiosemicarbazides 88(a-w) from the reaction of

isothiocyanate 87(a-c) and acid hydrazides 86(a-r) was confirmed by 1H NMR

Page 214: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

210

spectral data. Signals for NH proton of amide type linkage appeared in the range

of 10-11ppm and signals for two NH proton of thiourea type linkage appeared in

the range of 9-10 ppm. The 1H NMR data of a representativie

thiosemicarbazides 88m is given in the table 6.7. The formation of these

thiosemicarbazide were further confirmed from 13C NMR by the appearance of

new peaks in aromatic region and C=O peak in the range of 160-170ppm and

C=S peak in the range of 155-165ppm. The 13C NMR data of the

thiosemicarbazide 88m is given in the table 6.7

Table 6.7: 1H NMR and 13C NMR spectral data of Thiosemicarbazide 88m

HN

NH

S

NH

O

O

1 2

34

5

61′2′

3′4′5′

6′

OO

AB

1H NMR 13C NMR

Proton δ(ppm),intensity,multiplicity and coupling constants (J) Carbon δ(ppm)

NH 10.60 (s) C=S 181.5 NH 9.81 (s) C=O 166.3 NH 9.22 (s) ---- ---- H-1 ---- C-1 126.5 H-2 ---- C-2 152.6 H-3 7.19 (4H, m) C-3 111.9 H-4 7.19 (4H, m) C-4 126.1 H-5 7.19 (4H, m) C-5 120.3 H-6 7.19 (4H, m) C-6 128.2 H-1′ ---- C-1′ 134.8 H-2′ 6.93 (2H, t, J = 1.8 Hz) C-2′ 106.2 H-3′ ---- C-3′ 160.8 H-4′ 6.72 (1H, t, J = 2.1 Hz) C-4′ 104.2 H-5′ ---- C-5′ 160.8 H-6′ 6.93 (2H, t, J = 1.8 Hz) C-6′ 106.2

-OCH3 (B) 3.80 (6H, s) -OCH3 (B) 56.2 -OCH3 (A) 3.75(3H, s) -OCH3 (A) 55.9

Page 215: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

211

In mass fragmentation of 1-(3,5-dimthoxybenzoyl)-4-(2-methoxyphenyl)-

thiosemicarbazides 88m (Sheme 6.4), the molecular ion peak appeared at m/z

361 which confirmed the formation of 1-(3,5-dimthoxybenzoyl)-4-(2-

methoxyphenyl)thiosemicarbazides 88m and base peak appears at m/z 167.

.

HN

NH

S

NHO

O

O O

(m/z = 361, 12.4%)

S

NHO

(m/z = 167, 100 %)

OCN

(m/z = 133, 62.1 %)

O

(m/z = 108, 38.9 %)

HN

NH

SO

O O

(m/z = 240, 15.3 %)

O

O O

(m/z = 166, 35.3 %)

O O

(m/z = 138, 07.6 %)

HN

N NO

O

O O

HNNH2

O

O O

NHO

O O

(m/z = 196, 28.7%)

-SH2

-

-NCS-SNH2

O-

-NHNCS

-CO

HN

NH2O

O O

-NH2

SNH

O-

(m/z = 327, 4.4%)

(m/z = 180, 32.4%)

+

+

+

+

+ .+ .

+ .+ .

+ .

..

.

+

(88m)

Scheme 6.5: Mass fragmentation patern of thiosemicarbazide 88m

The elemental analysis of the 1-(3,5-dimthoxybenzoyl)-4-(2-

methoxyphenyl)thiosemicarbazides 88m, showed the best agreement of found

elemental percentage composition with calculated values as shown in table 6.8.

Table 6.8: Elemental analysis data of thiosemicarbazide 88m

Compd. % C H N S

Calculated 56.50 5.30 11.63 8.87 88m

Found 56.12 5.78 11.31 8.45

Page 216: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

212

The structure of 1-(3,5-dimthoxybenzoyl)-4-(2-methoxyphenyl)thiosemi-

carbazides 88m is finally confirmed by XRD analysis as explained below.

6.4.1 Crystal structure of 1-(3,5-Dimethoxybenzoyl)-4-(2-methoxyphenyl)thiosemicarbazide (88m)

Fig. 6.17: Crystal structure of 1-(3,5-Dimethoxybenzoyl)-4-(2-

methoxyphenyl)thiosemicarbazide (88m)

In the molecule of 1-(3,5-Dimethoxybenzoyl)-4-(2-methoxyphenyl)-

thiosemicarbazide (Fig. 6.17), the bond lengths and angles are in normal ranges.

The dihedral angle between the planar rings is 63.4 (2). The crystal structure of

(I) is stabilized by intramolecular N—-H···N and intermolecular N—H··· S

hydrogen bonds. Intermolecular N—H···S hydrogen bonds link the independent

molecules into dimers (Fig. 6.18). Dipole–dipole and van der Waals interactions

are also effective in the molecular packing in the crystal structure.

Page 217: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

213

Fig. 6.18: Packing diagram show the dipole–dipole and van der Waals

interactions are also in the crystal structure.

Crystal data C17H19N3O4S Dx = 1.371 Mg m−3 Mr = 361.41 Melting point: 435(2) K Monoclinic, P2 (1)/c Mo Kα radiation λ = 0.71073 Å a = 15.371 (6) Å Cell parameters from 1520 reflections b = 14.775 (6) Å θ = 2.7–24.9º c = 7.904 (3) Å µ = 0.21 mm−1

β = 102.835 (6) º T = 293 (2) K V = 1750.3 (12) Å3 Block, colorless Z = 4 0.46 × 0.26 × 0.20 mm F000 = 760

This Part of the chapter has been published: Ghulam Qadeer, Muhammad

Hanif, Nasim Hasan Rama and Wen-Tong Chen; Crystal structure of 1-(3′,5′-

Dimethoxybenzoyl)-4-(2′-methoxyphenyl)thiosemicarbazide; Acta Cryst. 2007,

E63, o3051.

Page 218: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

214

Geometric parameters (°A, °)

Selected bond lengths: S1—C13 1.6769 (17) C4—C5 1.383 (3) O1—C5 1.369 (2) N1—C6 1.415 (2)

C17—H17B 0.9600 C3—C4 1.381 (3) C17—H17C 0.9600 C3—H3A 0.9300

Selected bond angles: C5—O1—C15 117.66 (17) C8—C9—C10 119.24 (15) C8—O2—C16 118.15 (15) C8—C9—H9A 120.4 C10—O3—C17 117.66 (14) C10—C9—H9A 120.4 C13—N1—C6 130.71 (15)

O3—C10—C11 124.11 (16) C13—N1—H1B 114.6 C1—C6—C5 119.90 (17) H17A—C17—H17C 109.5 C9—C8—C7 120.89 (16) H17B—C17—H17C 109.5

6.4.2 Crystal structure of 1-[2-(2,4-Dichlorophenoxy)acetyl]-4-cyclohexylthiosemicarbazide 88d

Fig. 6.19: Crystal structure of 1-[2-(2,4-Dichlorophenoxy) acetyl]-4-

cyclohexylthiosemicarbazide 88d

Page 219: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

215

In 1-[2-(2,4-dichlorophenoxy)acetyl]-4-cyclohexylthiosemicarbazide (Fig. 6.19), the bond lengths and angles are in normal ranges. The thiosemicabazide

group is approximately planar and forms a dihedral angles of 88.03 (5)° with the

benzene ring. The crystal structure is stabilized by intermolecular N—-H···O and

N—H··· S hydrogen bonding. Dipole–dipole and van der Waals interactions are

also effective in the molecular packing in the crystal structure.

Crystal data

C15H19Cl2N3O2S F000 = 784 Mr = 376.29 Dx = 1.476 Mg m−3

Dm = 1.429 Mg m−3 Dm measured by not measured

Monoclinic, P21/c Melting point: 462(1) K Hall symbol: -P 2ybc Mo Kα radiation λ = 0.71073 Å a = 15.4180 (16) Å Cell parameters from 2164 reflections b = 12.1530 (13) Å θ = 2.1–27.5º c = 9.285 (1) Å µ = 0.52 mm−1 β = 103.299 (2) º T = 100 (2) K V = 1693.1 (3) Å3 Block, colorless Z = 4 0.25 × 0.20 × 0.15 mm

Geometric parameters (°A, °)

Selected bond lengths:

C1—O1 1.367 (3) C10—C11 1.511 (4) C1—C2 1.387 (4) C7—H7A 0.9900

C8—O2 1.222 (3) C8—N1 1.350 (3) N1—N2 1.382 (3) C10—N3 1.475

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Mohammad Azad Malik, Javeed Akhtar and James Raftery; Crystal

structure of 1-[2-(2,4-Dichloro-phenoxy)acetyl]-4-cyclohexylthiosemicarbazide,

Acta Cryst. 2007, E63, o3503.

Page 220: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

216

Selected bond angles: O1—C1—C2 125.0 (2) C12—C11—H11A 109.6 N1—C8—C7 115.0 (2)

C14—C15—H15B 109.6 N2—N1—H1 119.3 N3—C10—C15 110.4 (2)

Selected torsional bond angles: O1—C1—C2—C3 179.1 (2) C11—C12—C13—C14 55.5 (3) C6—C1—C2—C3 −0.5 (4) N2—C9—N3—C10 −176.2 O1—C1—C6—Cl2 1.4 (3)

S1—C9—N3—C10 4.4 (4) C2—C1—C6—Cl2 −178.9 C11—C10—N3—C9 87.9 (3) O1—C7—C8—O2 −173.6 C10—C11—C12—C13 −57.3 (3)

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N2—H2A···O2i 0.88 2.06 2.926 (3) 166 N3—H3A···S1ii 0.88 2.60 3.430 (2) 157

Symmetry codes: (i) x, −y+1/2, z+1/2; (ii) x, −y+1/2, z−1/2

6.4.3 Crystal structure of 1-[3-(4-methoxyphenyl) propanoyl]-4-(2-methoxyphenyl)thiosemicarbazide (88o)

Fig. 6.20: Crystal structure of 1-[3-(4-Methoxyphenyl)propanoyl]-4-(2-

methoxyphenyl)thiosemicarbazide 880

Page 221: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

217

Crystal data C18H21N3O3S Z = 4 Mr = 359.44 F000 = 760 Triclinic, P1 Dx = 1.322 Mg m−3 a = 9.0495 (16) Å Mo Kα radiation λ = 0.71073 Å b = 13.397 (2) Å Cell parameters from 3145 reflections c = 15.364 (3) Å θ = 2.3–26.3º α = 78.810 (3) º µ = 0.20 mm−1 β = 87.164 (3) º T = 100 (2) K γ = 81.269 (3) º Block, colourless V = 1805.7 (6) Å3 0.50 × 0.30 × 0.30 mm 6.4.4 Crystal structure of 1-(3,5-Difluorobenzoyl)-4-cyclohexyl-

thiosemicarbazide (88a)

Fig. 6.21: Crystal structure of 1-(3,5-Difluorobenzoyl)-4-

cyclohexylthiosemicarbazide 88a

Crystal data C16H23F2N3O2S2 Z = 6 Mr = 391.49 F000 = 1236 Hexagonal, P6 (1) Dx = 1.351 Mg m−3 a = 13.0740 (13) Å Mo Kα radiation λ = 0.71073 Å b = 13.0740 (13) Å Cell parameters from 1479 reflections

Page 222: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

218

c = 19.507 (4) Å θ = 3.3–19.3º α = 90º µ = 0.31 mm−1 β = 90º T = 100 (2) K γ = 120º Block, white V = 2887.6 (7) Å3 0.20 × 0.20 × 0.05 mm

Geometric parameters (°A, °)

Selected bond lengths: C11—H11A 0.9900 C2—H2 0.9500 C12—H12A 0.9900 C3—F1 1.361 (4) C12—H12B 0.9900

C3—C4 1.379 (5) C13—C14 1.552 (5) C4—C5 1.362 (5) C13—H13A 0.9900

Selected bond angles: C2—C1—C6 119.6 (4) C13—C12—C11 110.3 (4) C2—C1—C7 124.1 (4) C12—C13—H13A 109.5 N3—C9—C14 110.1 (3) S2—C16—H16A 109.5 N3—C9—C10 112.2 (3)

S2—C16—H16B 109.5 C14—C9—C10 110.6 (3) H16A—C16—H16B 109.5 C11—C10—C9 110.3 (3) H11A—C11—H11B 108.0 C15—S2—C16 98.2 (3)

Selected torsional bond angles: C6—C1—C2—C3 0.0 (6) C14—C9—C10—C11 57.6 (4) C7—C1—C2—C3 175.6 (4) F2—C5—C6—C1 −179.3 N3—C8—N2—N1 −4.3 (6) C2—C1—C6—C5 −0.4 (6)

S1—C8—N2—N1 179.4 (3) C7—C1—C6—C5 −176.3 C14—C9—N3—C8 −151.6 C6—C1—C7—N1 −170.9 C10—C9—N3—C8 84.8 (5) N3—C9—C10—C11 −179.1

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N1—H1···O2 0.88 1.91 2.769 (4) 165 N1—H1···S2 0.88 3.01 3.738 (3) 141 N2—H2A···O2i 0.88 2.15 2.882 (4) 140 N3—H3···O1ii 0.88 1.97 2.823 (4) 163

Symmetry codes: (i) x−y, x−1, z+1/6; (ii) y+1, −x+y+1, z−1/6

Page 223: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

219

6.5 Synthesis of Substituted 1,2,4-Triazol-3- thiones 89(a-t)

The respective 1,2,4-triazol-3-thiones 89(a-t) were synthesized by

refluxing the thiosemicarbazide in 4N aqueous sodium hydroxide solution307-308.

The products were purified by recrystallization from an appropriate solvent.

[Yield = 42 - 75%]

O

NH

HN

S

HN

R′

88(a-w)

X

RNaOH

NN

N SH

R′89(a-t)

X

R

Scheme 6.6: Synthesis of substituted Triazoles 89 (a-t)

89a. X = OCH2 R = 2,4-Cl2 R′ = -C6H11 89b. X = (CH2)2 R = 3,4,5-(OCH3)3 R′ = -C6H11 89c. X = 0 R = 3,5-F2 R′ = -C6H11 89d. X = 0 R = 3,5-(OCH3)2 R′ = -C6H11 89e. X = (CH2)2 R = 3,4,5-(OCH3)3 R′ = 2-MeO(C6H4)- 89f. X = 0 R = 3,5-(OCH3)2 R′ = 2-MeO(C6H4)- 89g. X = 0 R = 2,5-F2 R′ = 2-MeO(C6H4)- 89h. X = OCH2 R = 2,4-Cl2 R′ = 2-MeO(C6H4)- 89i. X = 0 R = 3,5-F2 R′ = 2-MeO(C6H4)- 89j. X = OCH2 R = 4-Br R′ = 2-MeO(C6H4)- 89k. X = (CH2)2 R = 4-OCH3 R′ = 2-MeO(C6H4)- 89l. X = O(CH2)2 R = 2,4-Cl2 R′ = 2-MeO(C6H4)- 89m. X = (CH2)2 R = 3,4,5-(OCH3)3 R′ = 4-MeO(C6H4)- 89n. X = 0 R = 3,5-(OCH3)2 R′ = 4-MeO(C6H4)- 89o. X = 0 R = 2,5-F2 R′ = 4-MeO(C6H4)- 89p. X = OCH2 R = 2,4-Cl2 R′ = 4-MeO(C6H4)- 89q. X = 0 R = 3,5-F2 R′ = 4-MeO(C6H4)- 89r. X = OCH2 R = 4-Br R′ = 4-MeO(C6H4)- 89s. X = (CH2)2 R = 4-OCH3 R′ = 4-MeO(C6H4)- 89t. X = O(CH2)2 R = 2,4-Cl2 R′ = 4-MeO(C6H4)-

Page 224: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

220

Mechanism

R NH

HN N

R'

O

S

OH-

H -H2O

R NH

HN N

R'

O

S-

NHHN

N SR

-O

R'H

OH

NHN

N SRHO

R'

H

-H2O

NHN

N SR

R' Scheme 6.7: Mechanism of dehydrative cyclization of Thiosemicarbazide in

basic medium

The physical and IR spectral data of compounds 88(a-t) is tabulated in table 6.9. The cyclization of thiosemicarbazides to 1,2,4--triazol-3-thiones was

indicated in the IR spectrum by the appearance of a single absorption for N–H

and disappearance of carbonyl absorption. In addition, a band appeared in the

region 1531-1481 cm-1 attributed to C=N group. The absorption bands in the

region of 1601-1411cm-1 were assigned to C=C absorptions of the aromatic ring.

Formation of the triazoles 89(a-t) was confirmed by 1H NMR. In 1H NMR

signals for N-H proton of triazoles appeared at the range of 13.0-14.5 ppm.

These protons gave broad signal and are highly deshielded. Disappearance of

other signals of N-H proton confirmed the formation of the triazoles. 1H NMR data

of the triazoles 89f is given in table 6.10. The formation of the triazoles 89(a-t) was also confirmed by disappearance of peak due to C=O in the range of 155-

165 ppm in 13C NMR spectrum. A new peak due to C=N appeared in the range of

158-168ppm. The 13C NMR spectral data of the triazole 89f is also given in the

table 6.10.

Page 225: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

221

Table 6.9: Physical and IR spectral data of 1,2,4-Triazoles 89(a-t)

νmax (cm-1) Compd. mp (°C) Yield *Rf

N-H Sp3 C-H

C=N C=C C=S

89a 178-180 58% 0.36 3409 3038-2873 1509 1591-1484 1271

89b 234-235 61% 0.31 3401 3032-2863 1501 1601-1471 1276

89c 221-223 67% 0.25 3411 3018-2853 1516 1589-1481 1255

89d 120-122 51% 0.36 3431 3090-2893 1521 1571-1454 1266

89e 218-219 54% 0.31 3403 3038-2873 1511 1601-1455 1256

89f 197-198 49% 0.35 3391 3108-2907 1501 1581-1474 1241

89g 133-134 75% 0.34 3361 3056-2940 1531 1584-1484 1244

89h 201-202 58% 0.40 3419 3081-2813 1498 1586-1456 1286

89i 169-170 61% 0.38 3307 3015-2903 1507 1588-1414 1247

89j 154-155 67% 0.36 3333 3101-2804 1513 1594-1384 1271

89k 204-205 51% 0.31 3291 3104-2903 1521 1571-1464 1265

89l 165-166 54% 0.35 3341 3054-2813 1491 1601-1484 1279

89m 169-170 56% 0.34 3249 3038-2933 1481 1551-1481 1257

89n 153-154 75% 0.39 3289 3031-2873 1497 1571-1464 1275

89o 173-175 58% 0.36 3381 3001-2874 1502 1601-1414 1245

89p 222-223 42% 0.31 3341 3018-2813 1514 1587-1411 1255

89q 229-230 66% 0.35 3349 3008-2803 1481 1571-1454 1248

89r 156-158 75% 0.34 3425 3108-2913 1521 1551-1435 1247

89s 102-103 58% 0.29 3401 3010-2870 1519 1601-1489 1276

89t 208-209 42% 0.28 3388 3038-2873 1505 1581-1444 1245

*Petroleum ether: acetone; (6:4)

Page 226: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

222

Table 6.10: 1H NMR and 13C NMR data of the triazole 89f

NNH

N

SO

O

1

23

4

5 6

1

23

4

56

O ′

′′

′ ′

A

B

Carbon 1H NMR 13C NMR

NH 14.07 (1H, s) ----

C=S ---- 169.41

C=N ---- 151.00

1 ---- 121.42

2 ---- 155.07

3 7.39(1H, dd, J = 7.8, 1.5Hz) 113.37

4 7.09(1H, ddd, J = 7.5, 1.2Hz) 123.63

5 7.49(1H, ddd, J = 9.0,1.5Hz) 121.42

6 7.17(1H, dd, J = 7.8, 0.6Hz) 128.00

1′ ---- 121.42

2′ 6.45(1H, d, J = 2.1Hz) 105.58

3′ ---- 160.66

4′ 6.52 (1H, dd, J = 2.4Hz) 102.76

5′ ---- 160.66

6′ 6.45(1H, d, J = 2.1Hz), 105.58

OCH3 (B) 3.78 (6H, s) 56.65

OCH3 (A) 3.63 (3H, s) 55.31

Page 227: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

223

In mass spectrum of 5-(3,5-Dimethoxyphenyl)-4-(2-methoxyphenyl)-4H-

1,2,4-triazole-3-thione 89f, the molecular ion peak appeared at m/z 343 which

confirmed the formation of 5-(3,5-Dimethoxyphenyl)-4-(2-methoxyphenyl)-4H-

1,2,4-triazole-3-thione 89f and base peak appeared at m/z 310 (Scheme 6.8)

NHN

NS O

O

O

(m/z = 343, 60.0 %)

NN

NHS O

O

O

NN

N O

O

O-SH

(m/z = 310, 100 %)

N

N O

O

O

NC

SH-

(m/z = 284, 06.7 %)

N

N O

O

(m/z = 177, 42.2 %)N

NO

O

O

(m/z = 178, 03.2 %)N

O

O

HN

NCSO

-H

(m/z = 163, 06.5 %)N

O

O

NO

O

N

N SHO

NC

SH-

NHO-H

N

N SO

(m/z = 148, 22.5 %)

(m/z = 180, 02.5 %)

(m/z = 122, 06.0 %)

(m/z = 179, 02.0 %)

O

+ +

+

++

.

.

.

.

.

+ . + .

+ .

+ .

+ . .

Scheme 6.8: Mass fragmentation pattern of Triazole 89f

Page 228: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

224

Finally the structure of 5-(3,5-Dimethoxyphenyl)-4-(2-methoxyphenyl)-4H-

1,2,4-triazole-3-thione 89f was confirmed by XRD analysis, details are given

below.

6.5.1 Crystal structure of 4-(2-methoxyphenyl)-5-(3,5-dimethoxy-

phenyl)-2H-1,2,4-triazole-3(4H)-thione (89f)

Fig. 6.22: Crystal structure of 4-(2-Methoxyphenyl)-5-(3,5-dimethoxyphenyl)-2H-

1,2,4-triazole-3(4H)-thione 89f

Page 229: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

225

The C1=S1 bond length [1.6782 (14) A °] compares with 1.6773 (19) A ° in

4-(4-chlorophenyl)-3-(furan-2-yl)-1H-1,2,4- triazole-5(4H)-thione309 and 1.668 (5)

A °in 4-amino-3-(1,2,3,4,5-pentahydroxypentyl)-1H-1,2,4-triazole-5(4H)-thione310.

In the triazole ring, the N2 ═C1 bond [1.3385 (17) A °] shows double-bond

character. In the crystal structure, all bond lengths and angles are comparable

with those observed in related structures309-310. The triazole ring is planar within

0.002 A °. the triazole group is almost idealy planar. 2-methoxyphenyl ring is

almost perpendicular and 3,5-dimethoxyphenyl ring is planar to the triazole ring.

It form inversion related dimers via N—H···S hydrogen bonds. The structure is

further stabilized by intermolecular-stacking interactions down the b axis. N2—

H2···S1 hydrogen bonds link molecules of title compound into infinite chains

extending along the b axis of the unit cell.

Crystal data C17H17N3O3S Z = 2 Mr = 343.40 F000 = 360 Triclinic, P1 Dx = 1.425 Mg m−3

Dm = 1.411 Mg m−3 Dm measured by not measured

Hall symbol: -P1 Melting point: 470(1) K a = 8.8950 (8) Å Mo Kα radiation λ = 0.71073 Å b = 9.3510 (8) Å Cell parameters from 3874 reflections c = 10.5510 (9) Å θ = 2.3–28.3º α = 94.3650 (10) º µ = 0.22 mm−1 β = 102.4520 (10) º T = 100 (2) K γ = 108.9240 (10) º Block, white V = 800.44 (12) Å3 0.35 × 0.30 × 0.25 mm

Geometric parameters (°A, °)

Selected bond lengths: C1—N2 1.3385 (17) C10—C11 1.3862 (19) C1—N1 1.3785 (16) C10—C15 1.4084 (18) C1—S1 1.6782 (14)

C16—O2 1.4337 (16) C5—H5 0.9500 N2—N3 1.3674 (15) C9—H9B 0.9800 N2—H2 0.8800

Page 230: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

226

Selected bond angles: N2—C1—N1 103.64 (11) C12—C11—H11 120.6 N2—C1—S1 128.55 (10) C12—C13—C14 118.86 (13) N1—C2—C10 128.40 (11)

C12—C13—H13 120.6 C8—C3—C4 121.02 (12) C14—C13—H13 120.6 C10—C11—H11 120.6 C14—O3—C17 116.86 (11)

Selected torsional bond angles: C8—C3—C4—O1 −179.51 (12) N1—C3—C4—O1 −2.65 (17) C4—C3—N1—C2 72.67 (17)

N1—C2—N3—N2 −0.04 (14) C15—C14—O3—C17 1.57 (19) C13—C14—C15—C10 0.75 (19)

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N2—H2···S1i 0.88 2.45 3.2624 (12) 154

Symmetry codes: (i) −x+1, −y+1, −z+1.

This Part of the chapter has been published: Ghulam Qadeer, Nasim Hasan

Rama, Javeed Akhtar, Mohammad Azad Malik, and James Raftery; Crystal

structure of 3-(3,5-dimethoxyphenyl)-4-(2-methoxyphenyl)-2H-1,2,4-triazole-

3(4H)-thione; Acta Cryst. 2007, E63, o3629.

Page 231: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

227

6.5.2 Crystal Structure of 5-(3,4,5-trimethoxyphenylethyl)-4-(2-

methoxyphenyl)-2H-1,2,4-triazole-3(4H)-thione (89e)

Fig. 6.23: Crystal Structure of 5-(3,4,5-Trimethoxyphenylethyl)-4-(2-

methoxyphenyl)-2H-1,2,4-triazole-3(4H)-thione 89e

Page 232: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

228

Crystal data C20H23N3O4S Z = 2 Mr = 401.47 F000 = 424 Triclinic, P1 Dx = 1.294 Mg m−3 Hall symbol: -P1 Melting point: 491(1) K a = 8.6368 (6) Å Mo Kα radiation λ = 0.71073 Å b = 10.5422 (7) Å Cell parameters from 5348 reflections c = 11.6944 (8) Å θ = 2.4–26.4º α = 91.7330 (10)º µ = 0.19 mm−1 β = 92.9550 (10)º T = 100 (2) K γ = 104.0750 (10)º Rectangular, colourless V = 1030.44 (12) Å3 0.55 × 0.35 × 0.30 mm

Geometric parameters (°A, °) Selected bond lengths: S1—C2 1.6775 (14) C7—C8 1.393 (2) O1—C7 1.3650 (18) C14—C19 1.3792 (19) N3—C1 1.3789 (18)

C1—C3 1.4892 (18) C16—C17 1.388 (2) C3—C4 1.5266 (19) C17—C18 1.384 (2) C4—C5 1.5122 (18)

Selected bond angles: C7—O1—C11 116.44 (13) C5—C10—H10 120.3 C8—O2—C12 113.01 (11) C9—C10—H10 120.3 C5—C6—C7 120.17 (13) C9—C8—C7 119.96 (12)

O4—C20—H20C 109.5 O3—C9—C10 124.48 (13) H20A—C20—H20C 109.5 C8—C9—C10 120.34 (13) H20B—C20—H20C 109.5 C5—C10—C9 119.45 (13)

Selected torsional bond angles: C1—N1—N2—C2 1.85 (15) C6—C7—C8—C9 −0.4 (2) N2—N1—C1—N3 −0.12 (15) C13—O3—C9—C8 −172.13 (13) N2—N1—C1—C3 −178.15 (13)

C2—N3—C14—C19 72.32 (18) C14—N3—C2—S1 3.5 (2) C1—N3—C14—C19 −107.68 (15) N1—C1—C3—C4 0.6 (2) C2—N3—C14—C15 −107.91 (15)

Page 233: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

229

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N2—H2N···O2i 0.878 (17) 1.890 (18) 2.7558 (15) 168.4 (15)

Symmetry codes: (i) x, y, z+1

6.5.3 Crystal Structure of 3-(4-Methoxyphenethyl)-4-(2-methoxy- phenyl)-1H-1,2,4-triazole-5(4H)-thione (89k)

Fig. 6.24: Crystal Structure of 3-(4-Methoxyphenethyl)-4-(2-methoxyphenyl)-1H-

1,2,4-triazole-5(4H)-thione 89k

Crystal data C18H19N3O2S Z = 4 Mr = 341.42 F000 = 720 Monoclinic, P21/n Dx = 1.279 Mg m−3 Hall symbol: -P 2yn Mo Kα radiation λ = 0.71073 Å a = 8.3664 (4) Å µ = 0.20 mm−1 b = 19.2172 (10) Å T = 293 (2) K

Page 234: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

230

c = 11.2800 (6) Å Block, colorless β = 102.1330 (10)º 0.38 × 0.30 × 0.24 mm V = 1773.07 (16) Å3

Geometric parameters (°A, °)

Selected bond lengths: C1—O1 1.431 (2) C10—H10A 0.9700 C1—H1A 0.9600 C10—H10B 0.9700 C1—H1B 0.9600

C11—H11B 0.9700 C2—C3 1.386 (2) C12—C17 1.365 (2) C2—C7 1.389 (2) C12—C13 1.382 (2)

Selected bond angles: O1—C1—H1A 109.5 C10—C11—H11A 109.5 O1—C1—H1B 109.5 C12—C11—H11B 109.5

H1A—C1—H1B 109.5 C14—C13—H13A 119.1 C12—C11—H11A 109.5

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N2—H1···S1i 0.823 (18) 2.468 (18) 3.2864 (14) 173.3 (19) C6—H6A···O2ii 0.93 2.57 3.278 (2) 133

Symmetry codes: (i) −x, −y+2, −z+2; (ii) −x+3/2, y+1/2, −z+3/2.

This Part of the chapter has been published: Ghulam Qadeer, Muhammad

Hanif, Nasim Hasan Rama and Wai-Yeung Wong; Crystal structure of 3-(4-

Methoxyphenethyl)-4-(2-methoxyphenyl)-1H-1,2,4-triazole-5(4H)thione; Acta

Cryst. 2007, E63, o3502.

Page 235: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

231

6.5.4 Crystal Structure of 3-(4-bromophenoxymethyl)-4-(4-methoxyphenyl)-1H-1,2,4-triazole-5(4H)-thione (89r)

Fig. 6.25: Crystal Structure of 3-(4-Bromophenoxymethyl)-4-(4-methoxyphenyl)-

1H-1,2,4-triazole-5(4H)-thione 89r

Crystal data C16H14BrN3O2S Z = 2 Mr = 392.27 F000 = 396 Triclinic, P1 Dx = 1.659 Mg m−3 a = 7.4590 (6) Å Mo Kα radiation λ = 0.71073 Å

Page 236: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

232

b = 7.7920 (7) Å Cell parameters from 3998 reflections c = 15.2870 (13) Å θ = 2.7–28.2º α = 97.3230 (10)º µ = 2.76 mm−1 β = 96.3740 (10)º T = 100 (2) K γ = 114.9470 (10)º Block, white V = 785.49 (12) Å3 0.20 × 0.20 × 0.15 mm

Geometric parameters (°A, °)

Selected bond lengths: Br1—C1 1.906 (2) C9—S1 1.677 (2) C1—C6 1.379 (3) C10—C11 1.378 (3)

N1—N2 1.374 (2) C9—N2 1.340 (3) N2—H2A 0.8800 C9—N3 1.379 (3)

Selected bond angles: C6—C1—C2 121.5 (2) C15—C10—N3 119.38 (18) C6—C1—Br1 118.95 (17) C10—C11—C12 119.71 (19)

C4—O1—C7 118.82 (16) C11—C10—C15 121.17 (19) C13—O2—C16 117.65 (17) C11—C10—N3 119.44 (18)

Selected torsional bond angles: C6—C1—C2—C3 −1.1 (3) N3—C8—N1—N2 −0.2 (2) Br1—C1—C2—C3 177.71 (17)

C7—C8—N1—N2 177.55 (19) C1—C2—C3—C4 0.2 (3) N3—C9—N2—N1 −0.3 (2)

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N2—H2A···S1i 0.88 2.37 3.2446 (18) 174

Symmetry codes: (i) −x, −y+2, −z+1

Page 237: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

233

6.5.5 Crystal Structure of 3-(2,4-dichlorophenoxymethyl)-4-(4-methoxyphenyl)-1H-1,2,4-triazole-5(4H)-thione (89p)

Fig. 6.26: Crystal Structure of 3-(2,4-Dichlorophenoxymethyl)-4-(4-

methoxyphenyl)-1H-1,2,4-triazole-5(4H)-thione 89p

Crystal data C16H13Cl2N3SO2 Z = 2 Mr = 382.64 F000 = 394 Triclinic, P1 Dx = 1.588 Mg m−3 a = 7.6529 (12) Å Mo Kα radiation λ = 0.71073 Å b = 7.6658 (12) Å Cell parameters from 1128 reflections c = 15.520 (3) Å θ = 2.9–27.4º α = 94.180 (3)º µ = 0.58 mm−1 β = 100.835 (3)º T = 100 (2) K γ = 113.856 (3)º Block, white V = 806.5 (2) Å3 0.23 × 0.12 × 0.10 mm

Page 238: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

234

Geometric parameters (°A, °)

Selected bond lengths C1—O2 1.375 (4) C9—S1 1.674 (3) C1—C6 1.389 (4) C10—C11 1.371 (4) C1—C2 1.396 (4) C10—C15 1.385 (4)

C10—N3 1.453 (4) C2—Cl1 1.731 (3) C11—C12 1.387 (4) C3—C4 1.387 (4) C11—H11 0.9500 C3—H3 0.9500

Selected bond angles C3—C2—Cl1 119.2 (3) C11—C12—C13 120.1 (3) C1—C2—Cl1 119.7 (2) C11—C12—H12 119.9 C2—C3—C4 118.3 (3) C13—C12—H12 119.9 C2—C3—H3 120.9

O1—C13—C14 115.3 (3) C4—C3—H3 120.9 O1—C13—C12 124.7 (3) C5—C4—C3 121.7 (3) C14—C13—C12 120.0 (3) C5—C4—Cl2 120.0 (3)

Selected torsional bond angles Cl1—C2—C3—C4 179.3 (2) C8—N1—N2—C9 0.4 (3) C2—C3—C4—C5 −1.9 (5) N2—C9—N3—C8 0.1 (3) C2—C3—C4—Cl2 177.6 (2) S1—C9—N3—C8 179.7 (2)

C3—C4—C5—C6 2.1 (5) S1—C9—N3—C10 0.6 (5) C4—C5—C6—C1 −0.5 (5) N1—C8—N3—C9 0.2 (4) O2—C1—C6—C5 179.2 (3) C2—C1—C6—C5 −1.3 (5)

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N2—H2···Cl3i 0.88 2.39 3.269 (3) 174

Symmetry codes: (i) −x+1, −y+1, −z

Page 239: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

235

6.6 Synthesis of thiadiazoles

Thiadiazoles 90(a-j) were synthesized by treating thiosemicarbazides with

concentrated H2SO4 at low temperature. Stirring continued over night and the

mixture was poured on crushed ice yielded thiadiazoles311. These thiadiazoles

were purified by recrystallization with suitable solvent. Physical data of these

thiadiazoles is given in table 6.11.

[Yield = 49 - 61%]

O

NH

HN

S

HN

R′

88(a-w)

X

RConc. H2SO4

NN

S NH

90(a-j)

X

RR′

Scheme 6.9: Synthesis of substituted Thidiazoles 90 (a-j)

90a. X = OCH2 R = 2,4-Cl2 R′ = -C6H11 90b. X = 0 R = 3,5-F2 R′ = -C6H11 90c. X = 0 R = 3,5-(OCH3)2 R′ = 2-OMe (C6H4)- 90d. X = 0 R = 3,4-(OCH3)2 R′ = 2-OMe (C6H4)- 90e. X = (CH2)2 R = 3,4,5-(OCH3)3 R′ = 2-OMe (C6H4)- 90f. X = O(CH2)2 R = 2,4-Cl2 R′ = 2-OMe (C6H4)- 90g. X = O(CH2)3 R = 2,4-Cl2 R′ = 2-OMe (C6H4)- 90h. X = OCH2 R = 2,4-Cl2 R′ = 2-OMe (C6H4)- 90i. X = O(CH2)3 R = 2,4-Cl2 R′ = 4-OMe (C6H4)- 90j. X = OCH2 R = 2,4-Cl2 R′ = 4-OMe (C6H4)-

Mechanism

R NH

O: HN

HN

SR'

H+

R NH

OHN

HN

SR'

+H

S

NHNNH

R'RHO

S

NHNNH

R'R

H2O

H+

+

-H2OS

NNNH

R'R

H

+-H+

S

NNNH

R'R

Scheme 6.10: Mechanism of dehydrative cyclization of the Thiosemicarbazides

in acidic medium

Page 240: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

236

Thiadiazoles 90(a-j) were characterized by IR spectra by disappearance

of peaks of thiourea type linkage of thiosemicarbazides. A peak due to N-H

stretch appeared in the range of 3300-3400 -1cm. A carbonyl peak of amide type

linkage also disappeared in the range of 1600-1680 -1cm. C=N stretch appeared

in the range of 1500-1510-1cm. The physical and IR data of thiadiazoles 90(a-j) is given in the table 6.11

Table 6.11: Physical and IR spectral data of Thiadiazoles 90(a-j)

νmax (cm-1) Compd

mp

(°C) Rf

Yield(%)

NH stretch

C-H stretch C=N

stretch C=C

90a 134-135 0.34 49 3301 3038-2873 1501 1601-1384

90b 198-199 0.38 51 3309 3021-2881 1511 1611-1385

90c 147-148 0.36 48 3291 3008-2821 1515 1591-1383

90d 189-191 0.39 58 3241 3018-2913 1521 1601-1414

90e 201-202 0.37 61 3315 3010-2893 1491 1615-1484

90f 178-179 0.35 59 3311 3039-2803 1508 1595-1384

90g 197-198 0.37 55 3206 3008-2870 1523 1594-1481

90h 188-189 0.35 57 3191 3031-2913 1503 1600-1484

90i 145-146 0.37 55 3299 3038-2945 1508 1594-1381

90j 111-112 0.37 54 3401 3058-2870 1531 1597-1394

1H NMR spectra confirmed the cyclization of thiosemicarbazide to

thiadiazole. In 1H NMR signal for N-H proton of thiadiazoles appeared at the

range of 13.0-14.5 ppm. These protons gave broad signals and were highly

deshielded. Disappearance of other signals of N-H proton confirmed the

formation of thiadiazoles. 1H NMR data of a representative thiadiazoles 90c is

given in table 6.12. The formation of thiadiazoles 90(a-j) was also confirmed by 13C NMR data. In 13C NMR, thiadiazoles were characterized by the

Page 241: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

237

disappearance of C=O peak of amide in the range of 165ppm. The 13C NMR data

of thiadiazole 90c is also given in the table 6.12.

Table 6.12: 1H and 13C NMR data of 2-Methoxyphenylamino-5-(3′,5′-

dimethoxyphenyl)-1,3,4-thiadiazole 90c

S

NN

O

O

HN

O1 2

34

56

123

4

5 6

′′′

′ ′

AB

Carbons 1H NMR 13C NMR

NH 8.97(1H, s) ----

C=N ---- 178.45

C=N ---- 155.27

1 ---- 134.09

2 ---- 150.84

3 7.29(1H, dd, J = 7.8, 2.0Hz) 116.58

4 7.05(1H, ddd, J = 8.1, 1.8Hz) 121.37

5 7.35(1H, ddd, J = 8.4,1.5Hz) 123.54

6 7.17 (1H, dd, J = 7.5, 1.6Hz) 116.58

1′ ---- 135.23

2′ 6.39(1H, d, J = 2.1Hz) 105.71

3′ ---- 165.54

4′ 6.59(1H, dd, J = 4.5, 2.4Hz) 99.71

5′ ---- 165.54

6′ 6.39(1H, d, J = 2.1Hz) 105.71

OCH3 (B) 3.77(6H, s) 56.54

OCH3 (A) 3.73(3H, s) 55.78

Page 242: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

238

In mass spectrum of 2-methoxyphenylamino-5-(3′,5′-dimethoxyphenyl)-

1,3,4-thiadiazole 90c, the molecular ion peak appeared at m/z 347 which

confirmed the formation of 2-methoxyphenylamino-5-(3′,5′-dimethoxyphenyl)-

1,3,4-thiadiazole 90c and base peak appeared at m/z 111 (Scheme 6.11).

N N

SNH

O

O Cl

(m/z = 347,10.0%)

N N

SNH2

O

O

(m/z = 237, 3.5%)

N N

SO

O

(m/z = 222, 2.5%)

-NH2

NS

NH2

NHO

O

(m/z = 74, 23.4%)

Cl

(m/z = 111,100%)

N

SO

O

(m/z = 195, 2%)

-N

SO

O

(m/z = 181, 31.2%)

N

SNH

Cl

(m/z = 184,10%)N

O

O

(m/z = 163, 36.3%)

O

O

(m/z = 137, 4.0%)

NO

O

Cl

CN

NC

O

O

S

NH(m/z = 195, 10.3%)

Cl

CNNH

(m/z = 152, 20.2%)

NCSO

O

-NCl

CHN

(m/z = 138, 21.5%)

+

++

++

-CN

.

..

.+ .

+ .

+ .+ .

+ .

+ .

+ .

+ .

Scheme 6.11: Mass fragmentation pattern of 2-Methoxyphenylamino-5-(3′,5′-

dimethoxyphenyl)-1,3,4-thiadiazole 90c

Page 243: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

239

6.7 Synthesis of indolinones

Indolinones 91(a-o) and 92(a-o) were synthesized by treating substituted

hydrazides with haloisatin in ethanol. These indolinones were purified by

recrystallization with suitable solvent. Physical data of these indolinones is given

in table 6.13.

[Yield = 79 - 91%]

X

R

NH

ONH2

NH

O

OR''

NH

O

NR''

HN

X

O

86(a-o) 91(a-ο)92(a-ο)

+

R″ = Cl, Br

C2H5OH

R

91a. X = 0 R = 3,5-F2 R″ = Cl 91b. X = 0 R = 2,5-F2 R″ = Cl 91c. X = 0 R = 2,6-F2 R″ = Cl 91d. X = 0 R = 3,4-(OCH3)2 R″ = Cl 91e. X = 0 R = 3,5-(OCH3)2 R″ = Cl 91f. X = 0 R = 2,4-(OCH3)2 R″ = Cl 91g. X = 0 R = 2,6-(OCH3)2 R″ = Cl 91h. X = CH2 R = 4-Cl R″ = Cl 91i. X = CH2 R = 4-F R″ = Cl 91j. X = (CH2)2 R = 3,4,5(OCH3)3 R″ = Cl 91k. X = (CH2)2 R = 4-OCH3 R″ = Cl 91l. X = OCH2 R = 4-Br R″ = Cl 91m. X = O(CH2)2 R = 2,4-Cl2 R″ = Cl 91n. X = O(CH2)3 R = 2,4-Cl2 R″ = Cl 91o. X = OCH2 R = 2,4-Cl2 R″ = Cl 92a. X = 0 R = 2,5-F2 R″ = Br 92b. X = 0 R = 3,5-F2 R″ = Br 92c. X = 0 R = 2,6-F2 R″ = Br 92d. X = 0 R = 3,4-(OCH3)2 R″ = Br 92e. X = 0 R = 3,5-(OCH3)2 R″ = Br 92f. X = 0 R = 2,4-(OCH3)2 R″ = Br 92g. X = 0 R = 2,6-(OCH3)2 R″ = Br 92h. X = CH2 R = 4-Cl R″ = Br 92i. X = CH2 R = 4-F R″ = Br

Page 244: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

240

92j. X = (CH2)2 R = 3,4,5(OCH3)3 R″ = Br 92k. X = (CH2)2 R = 4-OCH3 R″ = Br 92l. X = OCH2 R = 4-Br R″ = Br 92m. X = O(CH2)2 R = 2,4-Cl2 R″ = Br 92n. X = O(CH2)3 R = 2,4-Cl2 R″ = Br 92o. X = OCH2 R = 2,4-Cl2 R″ = Br

All the synthesized compounds were characterized by their physical

constants, IR (Table 6.13), 1H NMR, 13C NMR (Table 6.14), mass spectrometry

and X-ray crystallography. The IR spectra of indolinones exhibitied a strong

absorption in the region 1662-1618 cm-1 was assigned to the carbonyl group of

amide linkage, while absorption band for the secondary NH was observed in the

range 3476-3198 cm-1. The physical and IR spectral data of few indolinones are

given in table 6.13.

Table 6.13: Physical constants and IR spectral data of indolinones 91(a-i)

υmax (cm-1) S.No m.p (°C)

Yield (%)

NH strech

C-H strech

C=O C=N C=C

91a 178-179 89 3281 3084 1710 1677 1623, 1460 91b 165-166 81 3412 3115 1719 1670 1622, 1308 91c 212-213 84 3419 3118 1713 1678 1627, 1278 91d 145-146 70 3252 2936 1711 1674 1594, 1463 91e 112-113 85 3249 2845 1709 1670 1598, 1461 91f 201-202 80 3345 3138 1710 1675 1594, 1468 91g 155-156 85 3255 3132 1712 1670 1592, 1463 91h 139-140 89 3216 2853 1727 1691 1596, 1470 91i 188-189 76 3262 3149 1720 1678 1602, 1449

The formation of indolinones 91(a-o) and 92(a-o) from the respective

hydrazide and haloisatin was confirmed in the 1H NMR spectra by the

appearance of two NH proton signal in the region of 10.0- 12.0ppm. The

structures were futher confirmed by 13C NMR (Table 6.14). In 13C NMR, a signal

at δ 130-140ppm for C=N confirmed the formation of indolinones. The 1H NMR

Page 245: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

241

and 13C NMR of a representative indolinone 91a is given in table 6.14. The

detail of spectral data of other indolinones is given in chapter 7 (Experimental). Table 6.14: 1H NMR and 13C NMR spectral data of (Z)-N'-(5-Chloro-2-oxoindolin-

3-ylidene)-3,5-difluorobenzohydrazide 91a

O

NHN

F

F

NH

O

Cl

12

345

67

4a

7a

1

2

34

56

′′

′′

δ (ppm) and multiplicity Carbon

1H NMR 13C NMR

C=O ---- 171.3 2 ---- 164.2 3 ---- 136.7 4 8.11(1H, s) 127.1 4a ---- 116.8 5 ---- 127.4 6 7.68-7.43(4H, m) 132.8 7 6.93(1H, d, J = 8.7Hz) 126.0 7a ---- 143.3 1′ ---- 141.7

2′ 7.68-7.43(4H, m) 108.0 3′ ---- 164.7 4′ 7.68-7.43(4H, m) 112.2

5′ ---- 164.7 6′ 7.68-7.43(4H, m) 108.0

NH 10.98(1H, s) ----

Page 246: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

242

NH 11.78(1H, s) ----

In mass spectrum of (Z)-N'-(5-chloro-2-oxoindolin-3-ylidene)-3,5-difluoro-

benzohydrazide 91a, the molecular ion peak appeared at m/z 307 which

confirmed the formation of (Z)-N'-(5-chloro-2-oxoindolin-3-ylidene)-3,5-

difluorobenzohydrazide 91a and base peak appeared at m/z 194 (Scheme 6.12).

HN

O

NCl

HN

F

FO

-CO

NCl

HN

F

FO

NH

NCl

NHHN

O

NHN

HNCl

-CO

NHCl

NH

Cl

NH

Cl

(m/z = 335, 56.3%, Cl35)

+

(m/z = 307, 10.3%,Cl35)

+

++

+

+ +

(m/z = 166, 52.3%,Cl35)(m/z = 194, 100%,Cl35)

(m/z = 139, 15.3%,Cl35)(m/z = 152, 0.3%,Cl35)

(m/z = 75, 6.3%)(m/z = 115, 3.3%,Cl37)

(m/z = 309, 4.3%,Cl37)

(m/z = 337, 16.3%,Cl37)

(m/z = 168, 12.3%,Cl37)(m/z = 196, 33.2%,Cl37)

(m/z = 154, 0.3%,Cl37)

(m/z = 113, 55.3%,Cl35)

F

FO

F

FO

F

FO

(m/z = 141, 90.3%)

HN

O

NHN

Cl

(m/z = 131, 90.3%,Cl37)

+. +.

.

.

.

Page 247: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

243

Scheme 6.12: Mass fragmentation pattern of (Z)-N'-(5-Chloro-2-oxoindolin-3-

ylidene)-3,5-difluorobenzohydrazide (91a) Finally the structure of 91a was confirmed by XRD analysis. The

detail is given below.

6.7.1 Crystal Structure of (Z)-N'-(5-chloro-2-oxoindolin-3-ylidene) -3,5-difluorobenzohydrazide (90a)

Fig. 6.27: Crystal Structure of (Z)-N'-(5-Chloro-2-oxoindolin-3-ylidene)-3,5-

difluorobenzohydrazide (90a)

Crystal data C15H8ClF2N3O2 V = 1365.4 (2) Å3 Mr = 335.69 Z = 4 Monoclinic, P21/c Mo Kα a = 3.7932 (4) Å µ = 0.32 mm−1 b = 30.394 (3) Å T = 293 (2) K c = 11.8600 (11) Å 0.30 × 0.25 × 0.10 mm β = 93.034 (2)º

Geometric parameters (°A, °)

Page 248: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

244

Selected bond lengths C1—F1 1.356 (2) C9—O2 1.229 (2) C1—C2 1.359 (3) C9—N3 1.349 (3) C1—C6 1.377 (3) C10—C15 1.366 (3) C2—C3 1.392 (3)

C10—C11 1.400 (3) C2—H2A 0.9300 C10—N3 1.413 (3) C3—C4 1.389 (3) C11—C12 1.385 (3) C3—C7 1.491 (3) C12—C13 1.379 (3)

Selected bond angles C3—C2—H2A 120.5 C12—C11—C8 132.36 (19) C4—C3—C2 119.6 (2) C10—C11—C8 106.79 (17) C4—C3—C7 123.44 (19)

C12—C13—Cl1 119.54 (16) F2—C5—C6 117.9 (2) C14—C13—Cl1 118.77 (17) F2—C5—C4 118.23 (19) C15—C14—C13 120.8 (2)

Selected torsional bond angles C2—C1—C6—C5 −0.3 (4) C11—C10—C15—C14 −0.9 (3) N3—C10—C15—C14 178.6 (2)

C2—C3—C7—O1 9.1 (3) C11—C8—N2—N1 179.2 (2) C9—C8—N2—N1 −2.9 (3)

Hydrogen-bond geometry (Å, °) D—H···A D—H H···A D···A D—H···A N1—H1···O2 0.81 (2) 2.05 (2) 2.747 (3) 144 (2) N3—H2···O1i 0.86 (2) 2.17 (3) 2.890 (2) 142 (2) C15—H15A···O1i 0.93 2.44 3.130 (3) 131

Symmetry codes: (i) x+1, −y+1/2, z+1/2.

Page 249: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

245

Chapter-7 EXPERIMENTAL

7.1 Substrates and reagents Substituted carboxylic acids were products of Aldrich. Chloroform and

methanol were supplied by Lab Scan. Diethyl ether and dimethyl sulfoxide were

products of Riedel de Haёn. Magnesium sulphate was obtained from Fluka and

ethanol and ethyl acetate were obtained from commercial sources.

7.2 Purification of solvents All the solvents were used after necessary purification and drying

according to the standard procedures. The dried solvents were stored over

molecular sieves (4oA). A brief account of purification procedures employed is as

follow.

i. Methanol Calcium oxide (250g), freshly activated in a muffle furnace at 300-400 oC,

was introduced into a round bottom flask containing one litre of methanol. It was

refluxed for 6 hours and distilled at 68 oC.

ii. Absolute ethanol Calcium oxide (250g), freshly activated in a muffle furnace at 300-400 oC,

was introduced into a round bottom flask containing one litre of ethanol. It was

refluxed for 6 hours and then distilled at 77-78 oC.

iii. Acetone

Anhydrous calcium chloride (200g) was introduced into a round bottom

flask containing one litre of acetone and it was left for 4~5 hours. Pure acetone

was distilled at 56 oC.

Page 250: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

246

iv. Ethylacetate

In one litre of ethylacetate, 50ml of acetic anhydride and 10~15 drops of

conc.H2SO4 were added and refluxed for 5 hours. It was fractionated and treated

with 25g of potassium carbonate. It was filtered and distilled over 40g of calcium

hydride at 77 oC.

v. Diethyl ether

Diethyl ether was first distilled over anhydrous calcium chloride. The

distillate was refluxed on sodium wire, using benzophenone as an indicator.

When the colour of ether turned dark green, the mixture was distilled and stored

over molecular sieves (4 oA).

vi. Dimethyl sulphoxide

Calcium hydride (200g) was added to dimethyl sulfoxide (1000 ml) and

allowed to stand overnight. The solvent was filtered, fractionally distilled over

calcium hydride and stored on molecular sieves (4 oA).

7.3 Instrumentation The Rf-values were determined using pre-coated silica gel aluminium

packed plates Kiesal 60 F254 from Merck (Germany). Melting points of the

compounds were measured in open capillaries using Gallenkamp melting point

apparatus (MP-D) and are uncorrected. The IR spectra were recorded on FTS

3000 MX, Bio-Rad Merlin (Excalibur Model) spectrophotometer. The 1H-NMR

spectra were run on a Bruker 300 MHz NMR spectrometer in DMSO solution

using TMS as an internal standard. MS were recorded on Agilent technologies

6890N (GC) and an inert mass selective detector 5973 mass spectrometer.

Page 251: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

247

7.4 General procedure for the synthesis of esters

Substituted benzoic acids (0.2 moles) were dissolved in methanol/ethanol

(50 mL) in a round bottom flask equipped with a reflux condenser and a calcium

chloride guard tube. Concentrated sulfuric acid (2 mmoles) was added and the

reaction mixture subjected to reflux for 8-10 hours and monitored by thin layer

chromatography. After completion of the reaction, the excess alcohol was

removed under reduced pressure and resulting oil was poured into water. The

oily layer was separated and the aqueous portion extracted with diethyl ether (3 ×

50 mL). The combined organic layers were washed with dilute solution of sodium

carbonate (100 mL) to remove unreacted acid. The organic layer was dried over

anhydrous sodium sulphate. The solvent was removed on a rotary evaporator

after filtration.

Methyl 2,5-difluorobenzoate (85a): Yield: 92%; Oil; Rf: 0.77 (Petroleum ether :

ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3060 (sp2 CH Stretching), 2942 (sp3

CH Stretching), 1726 (C=O), 1585 (C=C), 1290 (C-O).

Methyl 3,5-difluorobenzoate (85b): Yield: 85%; Oil; Rf: 0.88 (Petroleum ether :

ethylacetate, 4:1)); IR (NaCl Cell, νmax, cm-1): 3058 (sp2 CH Stretching), 2940

(sp3 CH Stretching), 1727 (C=O), 1582 (C=C), 1309 (C-O). Methyl 2,6-difluorobenzoate (85c): Yield: 87%; Oil; Rf: 0.74 (Petroleum ether :

ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3061 (sp2 CH Stretching), 2943 (sp3

CH Stretching), 1728 (C=O), 1590 (C=C), 1313 (C-O). Methyl 3,4-dimethoxybenzoate (85d): Yield: 79%; m.p: 60-61 °C (lit. 59-62°C);

Rf: 0.57 (Petroleum ether : ethylacetate, 4:1); IR (KBr, νmax, cm-1): 3089 (sp2 CH

Stretching), 2901 (sp3 CH Stretching), 1740 (C=O), 1610 (C=C), 1304 (C-O).

Page 252: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

248

Methyl 3,5-dimethoxybenzoate (85e): Yield: 77%; m.p: 40-41°C (Lit. 41-43 °C);

Rf: 0.67 (Petroleum ether : ethylacetate, 4:1); IR (KBr, νmax, cm-1): 3063 (sp2 CH

Stretching), 2889 (sp3 CH Stretching), 1735 (C=O), 1625 (C=C), 1315 (C-O). Methyl 2,4-dimethoxybenzoate (85f): Yield: 80%; Oil; Rf: 0.76 (Petroleum ether

: ethylacetate, 4:1) (solvent for recrystallization: ethanol); IR IR (NaCl Cell, νmax,

cm-1): 3085 (sp2 CH Stretching), 2890 (sp3 CH Stretching), 1738 (C=O), 1596

(C=C), 1325 (C-O). Methyl 2,6-dimethoxybenzoate (85g): Yield: 81%; m.p: 87-88oC (Lit. 87-90oC)

[267]; Rf: 0.72 (Petroleum ether : ethylacetate, 4:1); IR (KBr, νmax, cm-1): 3030

(sp2 CH Stretching), 2989 (sp3 CH Stretching), 1725 (C=O), 1645 (C=C), 1296

(C-O).

Ethyl 2-(3-methoxyphenyl)acetate (85h): Yield: 78%; m.p: 125-126oC; Rf: 0.70

(Petroleum ether : ethylacetate, 4:1); IR (KBr, νmax, cm-1): 3056 (sp2 CH

Stretching), 2910 (sp3 CH Stretching), 1711 (C=O), 1569 (C=C), 1285 (C-O).

Methyl 2-(4-chlorophenyl)acetate (85i): Yield: 83%; Oil ; Rf: 0.71 (Petroleum

ether: ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3101 (sp2 CH Stretching),

3001 (sp3 CH Stretching), 1729 (C=O), 1578 (C=C), 1387 (C-O). Methyl 2-(4-flourophenyl)acetate (85j): Yield: 89%; Oil; Rf: 0.71 (Petroleum

ether : ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3048 (sp2 CH Stretching),

3010 (sp3 CH Stretching), 1725 (C=O), 1596 (C=C), 1269 (C-O). Methyl 3-(3,4,5-trimethoxyphenyl)propionate (85k): Yield: 79%; m.p: 88-90 oC;

Rf: 0.71 (Petroleum ether: ethylacetate, 4:1); IR (KBr, νmax, cm-1): 3084 (sp2 CH

Stretching), 2986 (sp3 CH Stretching), 1751 (C=O), 1586 (C=C), 1316 (C-O).

Page 253: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

249

Methyl 3-(4-methoxyphenyl)propionate (85l): Yield: 89%; m.p: 36-37 oC (Lit.

37-41oC); Rf: 0.71 (Petroleum ether : ethylacetate, 4:1); IR (KBr, νmax, cm-1): 3104

(sp2 CH Stretching), 2916 (sp3 CH Stretching), 1742 (C=O), 1610 (C=C), 1296

(C-O). Methyl 2-(4-bromophenoxy)acetate (85m): Yield: 82%; Oil ; Rf: 0.71

(Petroleum ether : ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3089 (sp2 CH

Stretching), 2986 (sp3 CH Stretching), 1726 (C=O), 1566 (C=C), 1310 (C-O). Methyl 2-(2,4-dichlorophenoxy)propionate (85n): Yield: 83%; Oil; Rf: 0.71

(Petroleum ether: ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3125 (sp2 CH

Stretching), 2896 (sp3 CH Stretching), 1722 (C=O), 1577 (C=C), 1296 (C-O).

Methyl 4-(2,4-dichlorophenoxy)butyrate (85o): Yield: 82%; Oil; Rf: 0.71

(Petroleum ether : ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3110 (sp2 CH

Stretching), 2975 (sp3 CH Stretching), 1745 (C=O), 1587 (C=C), 1283 (C-O). Methyl 2-(2,4-dichlorophenoxy)acetate (85p): Yield: 78%; m.p: Rf: 0.71

(Petroleum ether : ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3056 (sp2 CH

Stretching), 2974 (sp3 CH Stretching), 1720 (C=O), 1550 (C=C), 1300 (C-O).

Methyl 3-(4-methoxyphenyl)acrylate (85q): Yield: 89%; m.p: 86-87°C (lit. 88-

89°C); Rf: 0.71 (Petroleum ether : ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1):

3178 (sp2 CH Stretching), 2986 (sp3 CH Stretching), 1731 (C=O), 1612 (C=C),

1286 (C-O). Methyl 2-(2,4-dichlorophenylthio)acetate (85r): Yield: 87%; Oil; Rf: 0.71

(Petroleum ether : ethylacetate, 4:1); IR (NaCl Cell, νmax, cm-1): 3018 (sp2 CH

Stretching), 2946 (sp3 CH Stretching), 1735 (C=O), 1525 (C=C), 1310 (C-O).

Page 254: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

250

7.5 General procedure for the synthesis of hydrazides

The respective ester (0.02 moles) was dissolved in methanol (100 mL) in

a round bottom flask fitted with a reflux condenser and a calcium chloride drying

tube. Hydrazine hydrate (80%, 0.04 moles) was added slowly and the reaction

was monitored by thin layer chromatography. After completion of the reaction,

the reaction mixture was concentrated under reduced pressure. The resulting

crude solid was filtered, washed with water and recrystallized from aqueous

ethanol.

2,5-Difluorobenzohydrazide (86a): Yield: 78%; m.p: 141-142oC; Rf: 0.80

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3312 (NH stretching), 1626

(C=O), 1597 (C=C); 1H NMR (DMSO-d6, δ ppm ): 9.64 (1H, s, NH), 7.30-7.41

(3H, m, Ar-H), 4.58 (2H, s, NH2); 13C NMR (DMSO-d6, δ ppm ): 162.5(C=O),

[158.5, 155.3, 124.9, 119.1, 118.3, 116.5] (Ar-C); GCMS (DMF, m/z, %): 172 (22,

M+), 152 (3), 141 (100), 113 (52), 93 (4), 74 (2), 63 (20), 50 (5), 31 (6); Anal. Cald

for C7H6F2N2O : C, 48.84; H, 3.51; N, 16.27. Found: C, 48.71; H, 3.55; N, 16.22.

3,5-Difluorobenzohydrazide (86b): Yield: 74%; m.p: 158-159 oC; Rf: 0.87

(petroleum ether: acetone, 8:2); IR (KBr, νmax, cm-1): 3286 (NH stretching), 1628

(C=O), 1595 (C=C); 1H NMR (DMSO-d6, δ ppm ): 9.98 (1H, s, NH), 7.39-7.55

(3H, m, Ar-H), 4.60 (2H, s, NH2); 13C NMR (DMSO- d6, δ ppm ): 164.3 (C=O),

[163.5, 161.0, 137.1, 110.7), 110.5, 107.0] (Ar-C); GCMS (DMF, m/z, %): 172

(22, M+), 141 (100), 113 (71), 93 (4), 74 (2), 63 (29), 50 (5), 31 (6). Anal. Cald for

C7H6F2N2O : C, 48.84; H, 3.51; N, 16.27. Found: C, 48.81; H, 3.53; N, 16.23.

2,6-Difluorobenzohydrazide (86c): Yield: 88%; m.p: 244-246 oC; Rf: 0.75

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3316 (NH stretching), 1632

(C=O), 1579 (C=C); 1H NMR (DMSO-d 6, δ ppm ): 9.73 (1H, s, NH), 7.35-7.55

Page 255: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

251

(3H, m, Ar-H), 4.50 (2H, s, NH2); 13C NMR (DMSO-d6, δ ppm ): 164.0 (C=O),

[161.5, 161.0, 136.1, 112.7, 111.5, 109.0](Ar-C). GCMS (DMF, m/z, %):172

(11.02, M+), 141 (100), 113 (45.34), 93 (14.33), 74 (02.32), 63 (29.90), 50

(05.10), 31 (06.52). Anal. Cald for C7H6F2N2O : C, 48.84; H, 3.51; N, 16.27.

Found: C, 48.84; H, 3.43; N, 16.20.

3,4-Dimethoxybenzohydrazide (86d): Yield: 82%; m.p: 118 oC; Rf: 0.81

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3296 (NH stretching), 1661

(C=O), 1596 (C=C), 1255 (C-O). 1H NMR (DMSO-d 6, δ ppm ): 9.67 (1H, s, NH),

7.48 (1H, d, J = 2.1Hz, H-6 ), 7.45 (1H, d, J = 3Hz, H-2), 7.05 (1H, d, J = 8.4Hz,

H-5), 4.50 (2H, s, NH2) 3.79 (6H, s, 2 × OCH3); 13C NMR (DMSO-d 6, δ ppm ):

166.17 (C=O), [151.5, 148.6, 125.9, 120.5, 110.9, 105.2](Ar-C), 55.8 (OCH3).

GCMS (DMF, m/z, %): 196 (13, M+), 165 (100), 137 (22), 122 (13), 107 (8), 92

(5), 79 (12), 63 (4), 51 (7), 31 (2). Anal. Cald for C9H12N2O3 : C, 55.09; H, 6.16; N,

14.28; Found: C, 55.12; H, 6.13; N, 14.21.

3,5-Dimethoxybenzohydrazide (86e): Yield: 73%; m.p: 165-166oC; Rf: 0.78

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3410, 3325 (NH stretching),

1656 (C=O), 1578 (C=C), 1268 (C-O). 1H NMR (DMSO-d6, δ ppm ): 9.76 (1H, s,

NH), 6.99 (1H, d, J = 2.1 Hz, H-4), 6.62 (2H, dd, J = 3.0 Hz, H-2,6), 4.49 (2H, s,

NH2), 3.78 (6H, s, 2 × OCH3); 13C NMR (DMSO-d6, δ ppm ): 165.8 (C=O), [160.7,

160.5, 135.7, 105.2, 105.0, 103.4](Ar-C), 55.7 (OCH3). GCMS (DMF, m/z, %):196

(11, M+), 165 (100), 137 (32), 122 (23), 107 (18), 92 (35), 79 (12), 63 (14), 51

(27), 31 (5). Anal. Cald for C9H12N2O3 : C, 55.09; H, 6.16; N, 14.28; Found: C,

55.11; H, 6.23; N, 14.24.

2,4-Dimethoxybenzohydrazide (86f): Yield: 72%; m.p: 101-102oC; Rf: 0.80

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3350, 3210 (NH

stretching), 1668 (C=O), 1560 (C=C), 1256 (C-O). 1H NMR (DMSO-d6, δ ppm ):

9.69 (1H, s, NH), 7.77 (1H, d, J = 2.1 Hz, H-6 ), 6.67 (1H, d, J = 3.0 Hz, H-5),

6.41 (1H, d, J = 8.4 Hz, H-5), 4.72 (2H, s, NH2) 3.87 (6H, s, 2 × OCH3); 13C NMR

Page 256: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

252

(DMSO-d6, δ ppm ): 164.8 (C=O), 163.2, [159.2, 132.4, 113.3, 106.2, 98.9] (Ar-

C), 56.0 (OCH3). GCMS (DMF, m/z, %):196 (15, M+), 165 (100), 151 (39), 138

(19), 119 (43), 105 (10), 91 (15), 77 (18), 65 (4), 55 (4), 32 (3). Anal. Cald for

C9H12N2O3 : C, 55.09; H, 6.16; N, 14.28; Found: C, 55.05; H, 6.11; N, 14.19.

2,6-Dimethoxybenzohydrazide (86g): Yield: 79%; m.p: 160-161oC; Rf: 0.87

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3361, 3256 (NH

stretching), 1659 (C=O), 1589(C=C), 1254 (C-O). 1H NMR (DMSO- d 6, δ ppm ):

9.75(1H, s, NH), 6.95(1H, t, J = 7.1 Hz, H-4 ), 6.60(1H, d, J = 8.0 Hz, H-3,5),

4.51(2H, s, NH2), 3.75(6H, s, 2 × OCH3); 13C NMR (DMSO-d6, δ ppm ):

166.1(C=O), [160.9, 160.7, 135.7, 107.1, 105.5, 103.4](Ar-C), 55.8(OCH3).

GCMS (DMF, m/z, %): 196 (24, M+), 165 (100), 137 (14), 122 (23), 107 (8), 92

(5), 77 (22), 63 (4), 51 (7), 31 (2). Anal. Cald for C9H12N2O3 : C, 55.09; H, 6.16; N,

14.28; Found: C, 55.12; H, 6.13; N, 14.21.

2-(3-Methoxyphenyl)acetohydrazide (86h): Yield: 79%; m.p: 102-104oC; Rf:

0.75 (petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3396, 3241 (NH

stretching), 1654 (C=O), 1596 (C=C), 1256 (C-O); 1H NMR (DMSO- d 6, δ ppm ):

8.97 (1H, s, NH), 6.99 (1H, dd, J = 8.4, 2.1 Hz, H = 5’), 6.75-6.35 (3H, m, H = 2’,

4’,6’), 4.34 (2H, s, NH2), 3.84 (3H, s, OCH3), 3.29 (2H, s, CH2); 13C NMR (DMSO-

d 6, δ ppm ): 169.6 (C=O), [158.7, 135.5, 131.2, 121.5, 111.2](Ar-C), 56.3(OCH3),

40.7(CH2). GCMS (DMF, m/z, %): 180 (22, M+), 149 (25), 121 (100), 107 (12), 89

(44), 73 (6), 63 (20), 51 (5), 32 (64). Anal. Cald for C9H12N2O2 : C, 59.99; H, 6.71;

N, 15.55; Found: C, 59.92; H, 6.75; N, 15.51.

2-(4-Chlorophenyl)acetohydrazide (86i); Yield: 81%; m.p: 158-160oC; Rf: 0.81

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3385, 3274 (NH stretching),

1674 (C=O), 1574 (C=C); 1H NMR (DMSO- d 6, δ ppm ): 9.23 (1H, s, NH), 7.35

(2H, d, J = 8.4 Hz, H = 2’, 6’), 7.26 (2H, d, J = 8.4 Hz, H = 3’, 5’), 4.22 (2H, s,

NH2), 3.34 (2H, s, CH2); 13C NMR (DMSO- d 6, δ ppm ): 169.6 (C=O), [135.7,

Page 257: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

253

131.5, 131.2, 128.5](Ar-C), 40.7(CH2). GCMS (DMF, m/z, %): 184 (22, M+), 152

(35), 125 (100), 99 (12), 89 (44), 73 (6), 63 (20), 51 (5), 32 (64). Anal. Cald for

C8H9ClN2O : C, 52.04; H, 4.91; N, 15.17; Found: C, 52.09; H, 4.95; N, 15.11.

2-(4-Flourophenyl)acetohydrazide (86j): Yield: 78%; m.p: 121-122oC; Rf: 0.78

(petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3345, 3196 (NH

stretching), 1648 (C=O), 1585 (C=C); 1H NMR (DMSO-d6, δ ppm ): 9.25 (1H, s,

NH), 7.27 (2H, m, H = 2’, 6’), 7.10 (2H, m, H = 3’, 5’), 4.22 (2H, s, NH2), 3.39 (2H,

s, CH2); 13C NMR (DMSO-d 6, δ ppm ): 169.9 (C=O), [163.0, 132.8, 131.2, 131.1,

115.4, 115.2](Ar-C), 40.6(CH2); GCMS (DMF, m/z, %):168 (28, M+), 136 (37),

109 (100), 89 (2), 83 (24), 63 (10), 57 (15), 32 (34). Anal. Cald for C8H9FN2O : C,

57.14; H, 5.39; N, 16.66; Found: C, 57.04; H, 5.34; N, 16.61.

3-(3,4,5-Trimethoxyphenyl)propionatohydrazide (86k): Yield: 78%; m.p: 125-

126 oC; Rf: 0.80 (petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3341 (NH

stretching), 1659 (C=O), 1574 (C=C), 1248 (C-O). 1H NMR (DMSO-d 6, δ ppm ):

8.99 (1H, s, NH), 6.49 (2H, s, Ar-H), 4.20 (2H, s, NH2), 3.74 (9H, s, 3 × OCH3),

2.32 (2H, t, CH2), 2.75 (2H, t, CH2); 13C NMR (DMSO-d 6, δ ppm ): 171.3(C=O),

[153.14, 137.4, 136.0, 105.9](Ar-C), 56.16-(OCH3), 31.8 & 35.1(2 × CH2). GCMS

(DMF, m/z, %): 254 (68, M+), 223 (17), 207 (7), 181 (100), 165 (4), 148 (14), 136

(5), 121 (4), 105 (3), 91 (3), 77 ( 7), 65 (4). Anal. Cald for C12H8N2O4 : C, 56.68;

H, 7.13; N, 11.02; Found: C, 56.97; H, 7.11; N, 11.07.

3-(4-Methoxyphenyl)propionatohydrazide (86l): Yield: 74%; m.p: 110-112 oC;

Rf: 0.87 (petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3352 (NH

stretching), 1658 (C=O), 1572 (C=C), 1249 (C-O). 1H NMR (DMSO-d 6, δ ppm ):

8.98 (1H, s, NH), 7.09 (2H, d, J = 8.4 Hz, H-2’.6’), 6.86 (2H, dd, J = 1.8, 8.4 Hz,

H-3’.5’), 4.16 (2H, s, NH2), 3.39 (3H, s, OCH3), 2.50 (2H, t, CH2), 2.27 (2H, t,

CH2); 13C NMR (DMSO-d6, δ ppm ): 171.3 (C=O), [157.9,133.5, 129.5, 114.1](Ar-

C), 55.4 (OCH3), 30.6 & 35.8 (2 × CH2). GCMS (DMF, m/z, %): 194 (25), 162

Page 258: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

254

(14), 134 (10), 121 (100), 105 (03), 91(13), 77 ( 9), 65 (4). Anal. Cald for

C10H14N2O2 : C, 61.84; H, 7.27; N, 14.42; Found: C, 61.97'; H, 7.21; N, 14.37.

3-(4-Bromophenoxy)acetatohydrazide (86m): Yield: 82%; m.p: 164-165 oC; Rf:

0.75 (petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3345, 3210 (NH

stretching) 1654 (C=O), 1574(C=C), 1254 (C-O). 1H NMR (DMSO-d 6, δ ppm ):

9.39 (1H, s, NH), 7.43 (2H, d, J = 8.4 Hz, H-2’.6’), 6.92 (2H, d, J = 8.4 Hz, H-

3’.5’), 4.48 (2H, s, NH2) , 3.42 (2H, s, CH2); 13C NMR (DMSO-d 6, δ ppm ):

166.91 (C=O), [157.5, 132. 3, 117.2, 113.0](Ar-C), 66.7 (CH2). GCMS (DMF, m/z,

%): 246 [30, M+ (Br81)], 244 [(30, M+ (Br79)], , 185 & 187 (23), 172 & 174 (100),

155 & 157 (55), 143 & 145 (5), 117 & 119 (2), 73 & 75 (26), 63 & 65 (14), 45 (80).

Anal. Cald for C10H11BrN2O2 : C, 39.21; H, 3.70; N, 11.43; Found: C, 39.37'; H,

3.54 ; N, 11.37.

2-(2,4-Dichlorophenoxy)propionatohydrazide (86n): Yield: 78%; m.p: 154-155 oC; Rf: 0.81 (petroleum ether: acetone, 8:2); IR (KBr, νmax, cm-1): 3385, 3202 (NH

stretching) 1652 (C=O), 1547 (C=C), 1247 (C-O). 1H NMR (DMSO- d 6, δ ppm ):

9.35 (1H, s, NH), 7.57 (1H, d, J = 2.7 Hz, H-3’), 7.34 (1H, dd, J = 11.4, 2.4 Hz, H-

5’), 6.98 (1H, d, J = 9.0 Hz, H-6’), 4.75 (1H, q, CH), 4.32 (2H, s, NH2), 1.47 (3H,

d, CH3); 13C NMR (DMSO- d 6, δ ppm ): 169.6 (C=O), [152.4, 129.9, 128.4,

125.5, 123.5, 116.4](Ar-C), 74.30 (CH), 19.06 (CH3). GCMS (DMF, m/z, %): 250

(3, M+ (Cl37)), 248 (5, M+ (Cl35)), 191 (23, Cl37), 189 (40, Cl35), 164 (20 Cl37), 162

(32, Cl35), 154 (25), 145 (24), 133 (7), 125 (22), 109 (26), 98 (4), 87 (100), 75

(11), 59 (68). Anal. Cald for C9H10Cl2N2O2 : C, 43.40; H, 04.05; N, 11.25; Found:

C, 43.47; H, 04.02; N, 11.20.

2-(2,4-Dichlorophenoxy)butyratohydrazide (86o): Yield: 74%; m.p: 63-65 oC;

Rf: 0.78 (petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3374, 3216 (NH

stretching), 1663 (C=O), 1569 (C=C), 1257 (C-O). 1H NMR (DMSO-d 6, δ ppm ):

9.03 (1H, s, NH), 7.53 (1H, d, J = 2.1 Hz, H-3’), 7.33 (1H, dd, J = 2.4, 11.1 Hz, H-

5’), 7.13 (1H, d, J = 8.7 Hz, H-6’), 4.17 (2H, t, CH2), 4.03 (2H, s, NH2), 2.21 (2H,

Page 259: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

255

t, J = 7.5 Hz, CH2), 1.94 (2H, m, CH2). ); 13C NMR (DMSO-d 6, δ ppm ): 171.5

(C=O), [153.4, 129.6, 128.5, 124.7, 122.8, 115.4](Ar-C), [68.81, 30.06, 25.06]

(CH2). GCMS (DMF, m/z, %): 264 (2, M+, Cl37 ), 262 (2, M+

, Cl35), 233 (3, Cl37 ),

231 (2, Cl35 ), 177 (5, Cl37), 175 (2, Cl35), 164 (20, Cl37), 162 (32, Cl35), 147 (3,

Cl37), 145 (24, Cl35), 135 (3, Cl37), 133 (7, Cl35), 111 (4, Cl37), 109 (6, Cl35), 103

(4, Cl37), 101 (100, Cl35), 69 (11), 59 (68). Anal. Cald for C10H12Cl2N2O2 : C,

45.65; H, 4.60; N, 10.65; Found: C, 45.55; H, 4.63; N, 10.62.

2-(2,4-Dichlorophenoxy)acetohydrazide (86p): Yield: 88%; m.p: 151-152 oC;

Rf: 0.80 (petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3341, 3189 (NH

stretching), 1639 (C=O), 1547 (C=C), 1251 (C-O). 1H NMR (DMSO-d 6, δ ppm ):

9.26 (1H, s, NH), 7.53 (1H, d, J = 2.1 Hz, H-3’), 7.33 (1H, dd, J = 2.4, 11.1 Hz, H-

5’), 7.07 (1H, d, J = 8.7 Hz, H-6’), 4.61 (2H, s, CH2), 4.37 (2H, s, NH2); 13C NMR

(DMSO-d 6, δ ppm ): 166.4 (C=O), [153.0,129.7, 128.4, 125.4, 122.9, 115.6](Ar-

C), 67.5 (CH2); GCMS (DMF, m/z, %): 236 (12, M+, Cl37 ), 234 (32, M+

, Cl35), 201

(8, Cl37 ), 199 (22, Cl35 ), 177 (27, Cl35), 175 (35, Cl37), 164 (60, Cl37), 162 (92,

Cl35), 147 (33, Cl37), 145 (44, Cl35), 135 (8, Cl37), 133 (17, Cl35), 111 (44, Cl37),

109 (36, Cl35), 75 (34, Cl37), 73 (100, Cl35), 63 (21), 45 (88), 31 (29). Anal. Cald

for C8H8Cl2N2O2 : C, 40.88; H, 03.43; N, 11.92. Found: C, 40.73; H, 03.14; N,

11.82.

3-(4-Methoxyphenyl)acrylohydrazide (86q): Yield: 82%; m.p: 204-206 oC ; Rf:

0.87 (petroleum ether: acetone, 8:2); IR (KBr, νmax, cm-1): 3298, 3195 (NH

stretching), 1658 (C=O), 1601 (C=C), 1239 (C-O). 1H NMR (DMSO-d 6, δ ppm ):

8.98 (1H, s, NH), 7.09 (2H, d, J = 8.4 Hz, H-2’.6’), 6.86 (2H, d, J = 8.4, Hz, H-

3’.5’), 4.16 (2H, s, NH2), 3.39 (3H, s, OCH3), 2.50 (2H, d, J = 8.4, CH), 2.27 (2H,

d, J = 8.1, CH); 13C NMR (DMSO-d6, δ ppm ): 171.3 (C=O), 157.9, 143.9, 133.5,

129.5, 117.5, 114.1, 55.4 (OCH3); GCMS (DMF, m/z, %): 192 (21), 161 (10), 133

(21), 120 (100), 107 (1), 91 (13), 77 (9), 65(4). Anal. Cald for C10H12N2O2 : C,

62.49; H, 06.29; N, 14.57; Found: C, 62.97; H, 06.21; N, 14.37.

Page 260: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

256

2-(2,4-Dichlorophenylthio)acetohydrazide (86r): Yield: 72%; m.p: 60-62 oC; Rf:

0.75 (petroleum ether : acetone, 8:2); IR (KBr, νmax, cm-1): 3347 (NH stretching)

1645 (C=O), 1540 (C=C), 1281 (C-O). 1H NMR (DMSO-d 6, δ ppm ): 9.26 (1H, s,

NH), 7.53 (1H, d, J = 2.1 Hz, H-3’), 7.33 (1H, dd, J = 11.1, 2.4, Hz, H-5’), 7.07

(1H, d, J = 8.7 Hz, H-6’), 4.51 (2H, s, CH2), 4.37 (2H, s, NH2); 13C NMR (DMSO-

d 6, δ ppm ): 166.4 (C=O), [153.0, 129.7, 128.4, 125.4, 122.9, 115.6] (Ar-C),

43.5 (CH2); GCMS (DMF, m/z, %): 252 (10, M+, Cl37 ) 250 (12, M+

, Cl35), 217 (6,

Cl37 ), 215 (12, Cl35 ), 193 (17, Cl35), 191 (31, Cl37), 180 (20, Cl37), 178 (52, Cl35),

163 (53, Cl37), 161 (54, Cl35), 151 (8, Cl37), 149 (12, Cl35), 127 (44.36, Cl37), 125

(36.58, Cl35), 91 (34.32, Cl37), 89 (100, Cl35), 79 (21.24), 61 (88.36), 47 (29).

Anal. Cald for C8H8Cl2N2OS: C, 38.26; H, 03.21; N, 11.15; S, 12.77; Found: C,

38.06; H, 03.29; N, 11.11; S, 12.25.

7.6 General procedure for the synthesis of isothiocyanate

The substituted amine (0.25 moles) was dissolved in methanol (18 mL) in

a round bottom flask fitted with a reflux condenser. The whole assembly was

placed in an ice bath. Carbon disulfide (0.38 moles) and ammonia solution (1.0

mole) was added slowly. The temperature of the reaction mixture was maintained

at 15°C (not allowed to rise above 30°C) and stirred for 12h. After stirring, the

contents of the flask were transferred to a beaker. A solution of lead nitrate was

prepared in water prepared and added to the above mixture and stirred

overnight. The precipitates of lead sulphide formed along with isothiocyanate.

The isothiocyanate isolated by steam distillation. The isothiocyanate extracted by

using ethylacetate which was evaporated on rotary to get isothiocyanate. 2-Methoxyphenylisothiocyante (87a): Yield: 45%; Oily liquid; Rf: 0.82 (n-

hexane : ethyl acetate 8:2); IR (NaCl Cell, νmax, cm-1): 3069 (sp2 CH stretch),

2967, 2940 (sp3 CH), 2035b (N=C=S), 1591, 1495, 1457 (C=C); 1H NMR (CDCl3,

Page 261: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

257

δ ppm ): 7.31 (1H, d, J = 8.2Hz, Ar-H), 7.21 (1H, t, J = 7.9 Hz, Ar-H), 6.9 (1H, d, J

= 8.1 Hz, Ar-H ), 6.7 (1H, t, J = 8.3 Hz, Ar-H), 3.84 (3H, s, OCH3); 13C NMR (CDCl3, δ ppm ): 158.0 (C=S), [138.2, 130.5, 128.8, 123.1, 117.8, 113.3] (Ar-C),

54.5(OCH3); GC-MS(CHCl3, m/z.(%)): 165 (100), 150 (45), 132 (51), 122 (90), 78

(12), 63 (15), 51 (11), 45 (6), 39 (10); Anal. Cald for C8H7NOS: C, 58.16; H,

4.27; N, 8.48; S, 19.41; Found: C, 58.15; H, 4.20; N, 8.47; S, 19.49.

4-Methoxyphenylisothiocyante (87b): Yield: 44%; Oil; Rf: 0.81 (n-hexane :

ethyl acetate 8:2); IR (NaCl Cell, νmax, cm-1): 3053 (sp2 CH), 2957, 2907, 2836

(sp3 CH), 2106, b (N=C=S), 1603, 1503, 1460 (C=C); 1H NMR (CDCl3, δ ppm):

7.18 (2H, dt, J = 9.0, 3.3 Hz, Ar-H ), 6.86 (2H, dt, J = 9.0, 3.3 Hz, Ar-H), 3.80

(3H, s, OCH3); 13C NMR (CDCl3, δ ppm): 158.5(C=S), [133.8, 126.9,126.5, 123.5,

115.1, 114.8, 114.3] ( Ar-C), 55.5(OCH3); GC-MS (CHCl3, m/z, (%): 165 (100),

150(35), 132(59), 122(90), 82 (25 ), 78(25), 63(15), 51(11), 45(6), 39(10). Anal.

Cald for C8H7NOS: C, 58.16; H, 4.27; N, 8.48; S, 19.41; Found: C, 58.15; H,

4.20; N, 8.47; S, 19.49.

Cyclohexylisothiocyante (87c): Yield: 55%; Yellow oil; Rf: 0.68 (Petroleum

ether : ethyl acetate, 4:1); IR (KBr, νmax, cm-1): 2084, 2943-2970, 1454-1601. 1H

NMR (CDCl3, δ ppm ): δ 3.70-3.62(1H, m), 1.88-1.32(10H, m); 13C NMR (CDCl3):

δ 129.5(C=S), [55.3, 33.2, 24.9, 23.2](Cyclohexyl-H). Anal. Cald for C7H11NS: C,

59.53; H, 7.85; N, 9.92; S, 22.70; Found: C, 59.33; H, 7.84; N, 9.95; S, 22.77.

7.7 General procedure for the synthesis of Thiosemicarbazides

The substituted carboxylic acid hydrazide (6.8 mmoles) was dissolved in

methanol (30ml) and the substituted isothiocyanate (6.6 mmoles), separately

dissolved in methanol (10ml), was added dropwise with continuous stirring. The

reaction mixture was refluxed for 10-12 h. and monitored by TLC. After

consumption of the starting materials, the mixture was cooled to room

Page 262: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

258

temperature. Then, methanol was evaporated on rotary leaving behind crude

thiosemicarbazide as an oil that get solidified later on by cooling and

recrystallized from a mixture of ethylacetate and petroleum ether to yield

thiosemicarbazides 88(a-w).

1-(3,5-Difluorobenzoyl)-4-cyclohexylthiosemicarbazide (88a): Yield: 85%;

m.p: 131-133; Rf: 0.37 (Petroleum ether : acetone; 6:4); IR (KBr, νmax, cm-1):

3250-3145 (NH Stretching), 1653 (C=O), 1600, 1568 (C=C), 1265 (C=S); 1HNMR

(DMSO-d6, δ ppm ): δ 10.45 (1H, s, NH), 9.27 (1H, s, NH), 8.94 (1H, s, NH),

7.64-7.48 (3H, m, H-2,4,6), 4.13 (1H, m, H-1′), 1.78-1.04 (10H, m, cyclohexyl-H); 13C NMR (DMSO- d 6, δ ppm ): 164.0 (C=O), [162.7, 160.9, 136.4, 111.6, 108.0,

107.5] (Ar-C), [53.6, 36.2, 32.3, 31.2, 25.6, 25.4] (cyclohexyl-C); GCMS (DMF,

m/z, %): 313 (6, M+), 171 (100), 141 (13), 107 (8), 92 (5), 79 (12), 63 (4), 51 (7),

39 (2); Anal. Cald for C14H17F2N3OS : C, 53.66; H, 5.47; N, 13.41; S, 10.23;

Found: C, 53.61; H, 5.35; N, 13.44; S, 10.29.

1-[2-(2,4-Dichlorophenoxy)propanoyl]-4-cyclohexylthiosemicarbazide (88b):

Yield: 83%; m.p: 149-150°C; Rf: 0. 34 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3271-3149 (NH Stretching), 1659 (C=O), 1585 (C=C), 1251 (C=S); 1H

NMR (DMSO-d6, δ ppm ): 9.91 (1H, s, NH), 9.57 (1H, s, NH), 9.28 (1H, s, NH),

7.65-7.05 (3H, m, H-3′,5′,6′), 4.77 (1H, q, CH), 3.31 (1H, m, H-1), 1.79-1.07

(13H, m, cyclohexyl H); 13C NMR (DMSO-d6, δ ppm ): 167.0 (C=O), [153.9,

129.8, 128.3, 125.4, 122.8, 115.7] (Ar-C), 70.88 (CH), [53.3, 32.3, 25.6, 25.3]

(Cyclohexyl-C), 20.13 (CH3); GCMS (DMF, m/z, %): 390 (3, M+), 247 (100), 217

(18), 172 (43), 144 (24), 141 (13), 111 (59), 83 (18), 60 (11), 79 (12), 63 (4), 51

(7), 39 (2); Anal. Cald for C16H21Cl2N3O2S : C, 49.23; H, 5.42; N, 10.77; S, 8. 21;

Found: C, 49.27; H, 5.45; N, 10.73; S, 8. 28.

1-(3,4-Dimethoxybenzoyl)-4-cyclohexylthiosemicarbazide (88c): Yield: 80%;

m.p: 185-186; Rf: 0.38 (Petroleum ether : acetone; 6:4); IR (KBr, νmax, cm-1):

Page 263: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

259

3351-3115 (NH Stretching), 1666 (C=O), 1561 (C=C), 1239 (C=S); 1H NMR

(DMSO-d 6, δ ppm ): 10.05 (1H, s, NH), 9.37 (1H, s, NH), 8.84 (1H, s, NH), 7.54-

7.41 (3H, m, H-2,5,6), 4.10 (1H, s, H-1′), 1.71-1.09 (10H, m, cyclohexyl-H), 3.71

(6H, s, OCH3); 13C NMR (DMSO-d 6, δ ppm ): 166.0 (C=O), [158.7, 155.9, 136.4,

121.6, 118.0, 111.5] (Ar-C), 55.8 (OCH3), [53.6, 36.2, 31.3, 31.2, 25.6, 25.4]

(Cyclohexyl-C); GCMS (DMF, m/z, %): 337 (3, M+), 247 (100), 217 (18), 172

(43), 144 (24), 141 (13), 111 (59), 83 (18), 60 (11), 79 (12), 63 (4), 51 (7), 39 (2); Anal. Cald for C16H23N3O3S : C, 56.95; H, 6.87; N, 12.45; S, 9.50; Found: C,

56.93; H, 6.81; N, 12.49; S, 9.55.

1-[2-(2,4-Dichlorophenoxy)acetyl]-4-cyclohexylthiosemicarbazide (88d):

Yield: 79%; m.p: 189-190°C; Rf: 0.35 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3290-3009 (NH Stretching), 1654 (C=O), 1560 (C=C), 1255 (C=S); 1H

NMR (DMSO-d 6, δ ppm ): 9.95 (1H, s, NH), 9.47 (1H, s, NH), 9.18 (1H, s, NH),

7.62-7.08 (3H, m, H-3′,5′,6′), 4.74 (2H, s, CH2), 3.07 (1H, s, H-1), 1.79-1.07

(10H, m, cyclohexyl-H); 13C NMR (DMSO-d6, δ ppm ): 167.0 (C=O), [152.9,

129.8, 128.3, 125.4, 122.8, 115.7] (Ar-C), 66.88 (CH2), [53.3, 32.3, 25.6, 25.3]

(Cyclohexyl-C). GCMS (DMF, m/z, %): 376 (21, M+), 232 (100), 230 (14), 202

(2), 172 (11), 144 (42), 141 (22), 111 (13), 77 (8), 63 (4), 51 (7); Anal. Cald for

C15H19Cl2N3O2S : C, 47.88; H, 5.09; N, 11.17; S, 8.52; Found: C, 47.83; H, 5.01;

N, 11.14; S, 8.62.

1-[3-(3,4,5-Trimethoxyphenyl)propanoyl]-4-cyclohexylthiosemicarbazide (88e): Yield: 84%; m.p: 174-176; Rf: 0.37 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3400-3145 (NH Stretching), 1658 (C=O), 1598 (C=C), 1244

(C=S); 1H NMR (DMSO-d6, δ ppm ): 10.17 (1H, s, NH), 9.64 (1H, s, NH), 9.04

(1H, s, NH), 7.91 (1H, d, J = 2.1 Hz, H-6′), 7.76 (1H, d, J = 2.1 Hz, H-2′), 4.10

(1H, s, H-1), 3.73 (9H, s, 3 × OCH3), 2.81 (2H, t, J = 7.5 Hz, CH2), 2.51 (2H, t, J

= 8.1 Hz, CH2), 1.71-1.09 (10H, m, cyclohexyl-H); 13C NMR (DMSO-d 6, δ ppm ):

180.5 (C=S), 176.2 (C=O), [152.6, 136.2, 132.1, 105.9, 105.8] (Ar-C), 56.15

Page 264: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

260

(OCH3), [53.6, 36.2, 35.4, 31.3, 31.3, 31.2, 25.6, 25.4] (Cyclohexyl & alphitic-C);

GCMS (DMF, m/z, %): 395 (3, M+), 253 (100), 238 (22), 195 (11), 157 (21), 141

(13), 107 (9), 92 (5), 79 (12), 63 (4), 51 (7); Anal. Cald for C19H29N3O4S : C,

57.70; H, 7.39; N, 10.92; S, 8.11; Found: C, 57.77; H, 7.32; N, 10.95; S, 8.17.

1-[3-(4-Methoxyphenyl)propanoyl]-4-cyclohexylthiosemicarbazide (88f): Yield: 85%; m.p: 133-135; Rf: 0.33 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3411-3205 (NH Stretching), 1673 (C=O), 1588 (C=C), 1242 (C=S);

1H NMR (DMSO-d6, δ ppm ): 10.87 (1H, s, NH), 9.77 (1H, s, NH), 9.53 (1H, s,

NH), 7.29 (2H, d, J = 8.4 Hz, H-2′,6′), 7.17 (2H, d, J = 8.4 Hz, H-3′,5′), 3.74 (3H,

s, OCH3), 2.65 (2H, t, CH2), 2.51 (1H, m, CH2), 2.33 (1H, m, H-1), 1.79-1.19

(10H, m, cyclohexyl-H); 13C NMR (DMSO-d 6, δ ppm ): 169.7 (C=O), [156.9,

133.4, 132.3, 129.6, 114.2] (Ar-C), 55.61 (OCH3), [53.6, 36.2, 35.7, 31.3, 31.2),

29.9, 25.6, 25.4] (Cyclohexyl & aliphetic-C); GCMS (DMF, m/z, %): 335 (7, M+),

301 (3), 237 (21), 193 (100), 178 (12), 163 (15), 141(52), 135 (8), 112 (10), 99

(5), 83 (9); Anal. Cald for C17H25N3O2S : C, 60.87; H, 7.51; N, 12.53; S, 9.56;

Found: C, 60.75; H, 7.57; N, 12.45; S, 9.52.

1-[2-(4-Bromophenoxy)acetyl]-4-(2-methoxyphenyl)thiosemicarbazide (88g): Yield: 80%; m.p: 141-142oC; Rf: 0.34 (petroleum ether : acetone, 6:4); IR

(KBr, νmax, cm-1): 3385-3216 (NH Stretching), 1685 (C=O), 1605 (C=C), 1255

(C=S); 1H NMR (DMSO-d 6, δ ppm ): 9.95 (1H, s, NH), 9.69 (1H, s, NH), 9.32

(1H, s, NH), 7.81-6.65 (8H, m, Ar-H), 4.51 (2H, s, H-1), 3.79 (3H, s, OCH3); 13C

NMR (DMSO-d 6, δ ppm ): 167.1 (C=O), [158.4, 157.6, 133.7, 128.5, 128.3,

123.4, 118.8, 111.8] (Ar-C), 68.55 (CH2), 56.6 (OCH3); GCMS (DMF, m/z, %): 410(19, M+), 377 (2), 287 (25), 244 (100), 228 (15), 213 (45), 185 (11), 165

(100), 133 (2), 123 (13), 107 (8); Anal. Cald for C16H16BrN3O3S : C, 46.84; H,

3.93; N, 10.24; S, 7.82; Found: C, 46.89; H, 3.97; N, 10.19; S, 7.89.

1-(2,5-Difluorobenzoyl)-4-(2-methoxyphenyl)thiosemicarbazide (88h): Yield:

78%; m.p: 153-154oC; Rf: 0.37 (petroleum ether : acetone, 6:4); IR (KBr, νmax,

Page 265: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

261

cm-1): 3285-3210 (NH Stretching), 1670 (C=O), 1605 (C=C), 1245 (C=S); 1H

NMR (DMSO-d 6, δ ppm ): 10.62 (1H, s, NH), 9.92 (1H, s, NH), 9.19 (1H, s, NH),

7.96-7.52 (3H, m, H-3′,4′,6′), 7.18 (1H, dd, J = 7.5 Hz, H-5), 7.06 (1H, d, J = 7.8

Hz, H-3), 6.94 (1H, dd, J = 7.8 Hz, H-4), 6.73 (1H, d, J = 7.8 Hz, H-6), 3.79 (3H,

s, OCH3); 13C NMR(DMSO-d 6, δ ppm ): 165.3 (C=O), [160.8, 153.6, 129.8,

126.2, 123.5, 120.2, 115.9] (Ar-C), 55.1 (OCH3); GCMS (DMF, m/z, %): 337 (11,

M+), 303 (11), 215 (2), 172 (5), 165 (100), 156 (5), 141(54), 133 (14), 123 (19),

113 (8), 107 (21); Anal. Cald for C15H13F2N3O2S : C, 53.41; H, 3.88; N, 12.46; S,

9.91; Found: C, 53.49; H, 3.79; N, 12.44; S, 9.98.

1-[2-(2,4-Dichlorophenoxy)acetyl]-4-(2-methoxyphenyl)thiosemicarbazide (88i): Yield: 85%; m.p: 156-157oC; Rf: 0.40(petroleum ether : acetone, 6:4); IR

(KBr, νmax, cm-1): 3385-3216 (NH Stretching), 1671 (C=O), 1586 (C=C), 1248

(C=S); 1H NMR (DMSO-d6, δ ppm ): 10.05 (1H, s, NH), 9.61 (1H, s, NH), 9.02

(1H, s, NH), 7.88-6.89 (7H, m, Ar-H), 4.11 (2H, s, CH2), 3.76 (3H, s, OCH3); 13C

NMR (DMSO-d 6, δ ppm ): 169.1(C=O), [153.4, 151.6, 129.7, 128.5, 128.5,

126.4, 124.8, 122.8, 120.2, 115.5, 111.8] (Ar-C), 68.75 (CH2), 56.1 (OCH3); GCMS (DMF, m/z, %): 400 (3, M+), 365 (20), 277 (28), 234 (56), 218 (4), 203

(23), 175 (19), 165 (100), 133 (2), 123 (12), 107 (8); Anal. Cald for

C16H15Cl2N3O3S : C, 48.01; H, 3.78; N, 10.50; S, 8.01; Found: C, 48.11; H, 3.74;

N, 10.51; S, 8.07.

1-(3,5-Difluorobenzoyl)-4-(2-methoxyphenyl)thiosemicarbazide (88j): Yield:

83%; m.p: 149-150oC; Rf: 0.39 (petroleum ether : acetone, 6:4); IR (KBr, νmax,

cm-1): 3315-3116 (NH Stretching), 1645 (C=O), 1587 (C=C), 1265 (C=S); 1H

NMR (DMSO-d 6, δ ppm ): 10.52 (1H, s, NH), 9.72 (1H, s, NH), 9.39 (1H, s, NH),

7.83-7.22 (7H, m, Ar-H), 3.84 (3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ):

165.3 (C=O), [160.5, 155.3, 129.8, 127.2, 123.5, 120.2, 115.9, 111.9] (Ar-C),

56.1 (OCH3); GCMS (DMF, m/z, %): 337 (29, M+), 303 (2), 215 (12), 172 (15),

165 (100), 156 (9), 141 (50), 133 (15), 123 (21), 113 (51), 107 (25); Anal. Cald

Page 266: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

262

for C15H13F2N3O2S : C, 53.41; H, 3.88; N, 12.46; S, 9.91; Found: C, 53.47; H,

3.79; N, 12.42; S, 9.93.

1-(3,4-Dimethoxybenzoyl)-4-(2-methoxyphenyl)thiosemicarbazide (88k): Yield: 80%; m.p: 173-175oC; Rf: 0.41 (petroleum ether : acetone, 6:4); IR (KBr,

νmax, cm-1): 3410-3016 (NH Stretching), 1660 (C=O), 1587 (C=C), 1251(C=S); 1H

NMR (DMSO-d 6, δ ppm ): 10.25 (1H, s, NH), 9.89 (1H, s, NH), 9.42 (1H, s, NH),

7.79 (1H, dd, J = 8.1, 2.1 Hz, H-6′), 7.21-7.05 (4H, m, H-3,4,5,6), 6.95 (1H, d, J

= 2.4 Hz, H-2′), 6.72 (1H, d, J = 8.1Hz, H-5′), 3.88 (6H, s, OCH3), 3.71 (3H, s,

OCH3); 13C NMR (DMSO-d 6, δ ppm ): 166.5 (C=O), [160.3, 153.6, 133.8, 128.3,

126.5, 120.2, 111.9, 106.1, 104.2] (Ar-C), 56.1-55.9 (OCH3); GCMS (DMF, m/z,

%): 361 (24, M+), 327 (25), 239 (20), 196 (19), 180 (26), 165 (100), 165 (21), 137

(12), 123 (13), 107 (8); Anal. Cald for C17H19N3O4S : C, 56.50; H, 5.30; N, 11.63;

S, 8.87; Found: C, 56.60; H, 5.33; N, 11.50; S, 8.85.

1-(2,6-Dimethoxybenzoyl)-4-(2-methoxyphenyl)thiosemicarbazide(88l): Yield: 79%; m.p: 180-182oC; Rf: 0.35 (petroleum ether : acetone, 6:4); IR (KBr,

νmax, cm-1): 3400-3260 (NH Stretching), 1663 (C=O), 1625, 1541 (C=C), 1261

(C=S); 1H NMR (DMSO-d6, δ ppm ): 10.65 (1H, s, NH), 9.71 (1H, s, NH), 9.35

(1H, s, NH), 7.79 (1H, dd, J = 8.4Hz, H-4′), 7.63 (1H, dd, J = 8.1Hz, H-5), 7.01-

6.89 (4H, m, H-3,4,3′,5′), 6.62 (1H, d, J = 8.1Hz, H-6), 3.95 (6H, s, OCH3), 3.70

(3H, s, OCH3); 13C NMR (DMSO-d 6, δ ppm ): 164.5 (C=O), [158.4, 158.0, 135.5,

128.2, 126.3, 121.2, 111.9, 106.1](Ar-C), 56.22-55.74(OCH3); GCMS (DMF, m/z,

%): 361 (6, M+), 327 (20), 239 (33), 196 (92), 180 (26), 165 (100), 137 (12), 123

(13), 107 (8); Anal. Cald for C17H19N3O4S : C, 56.50; H, 5.30; N, 11.63; S, 8.87;

Found: C, 56.54; H, 5.29; N, 11.62; S, 8.78.

1-(3,5-Dimethoxybenzoyl)-4-(2-methoxyphenyl)thiosemicarbazide(88m): Yield: 84%; m.p: 165-167oC; Rf: 0.37 (petroleum ether : acetone, 6:4); IR (KBr,

νmax, cm-1): 3305-3011 (NH Stretching), 1667 (C=O), 1561 (C=C), 1250 (C=S); 1H

Page 267: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

263

NMR (DMSO-d 6, δ ppm ): 10.60 (1H, s, NH), 9.81 (1H, s, NH), 9.22 (1H, s, NH),

7.90 (1H, d, J = 2.4Hz, H-2′,6′), 7.19-7.02 (4H, m, H-3,4,5,6), 6.72 (1H, dd, J =

2.1 Hz, H-4′), 3.80 (6H, s, OCH3), 3.75 (3H, s, OCH3); 13C NMR (DMSO-d 6, δ

ppm ): 166.3 (C=O), [160.8, 152.6, 134.8, 128.2, 126.5, 120.2, 111.9, 106.1,

104.2] (Ar-C), 56.1-5.9 (OCH3); GCMS (DMF, m/z, %): 361(12, M+), 327 (41),

239 (02), 196 (10), 180 (26), 165 (100), 165 (21), 137 (12), 123 (13), 107 (8); Anal. Cald for C17H19N3O4S : C, 56.50; H, 5.30; N, 11.63; S, 8.87; Found: C,

56.59; H, 5.34; N, 11.53; S, 8.81.

1-[3-(3,4,5-Trimethoxyphenyl)propanoyl]-4-(2-methoxyphenyl)thiosemi- carbazide (88n): Yield: 85%; m.p: 174-175oC; Rf: 0.40 (petroleum ether :

acetone, 6:4); IR (KBr, νmax, cm-1): 3445-3211 (NH Stretching), 1656 (C=O), 1525

(C=C), 1255 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 10.07 (1H, s, NH), 9.74 (1H, s,

NH), 8.97 (1H, s, NH), 7.91 (1H, s, H-6′), 7.16-6.97 (4H, m, H-3,4,5,6), 6.55 (1H,

s, H-2′), 3.79 (9H, s, OCH3), 3.62 (3H, s, OCH3), 2.85 (2H, t, J = 7.5 Hz, CH2),

2.54 (2H, t, J = 8.1 Hz, CH2); 13C NMR (DMSO-d6, δ ppm ): 180.9 (C=S),

176.5(C=O), [153.2, 152.6, 137.2, 136.1, 128.1, 127.3, 126.5, 120.2, 111.8,

105.9, 105.8] (Ar-C), 60.3-56.1 (OCH3), 35.4 (CH2), 31.3 (CH2); GCMS (DMF,

m/z, %): 419 (9, M+), 385 (2), 254 (10), 238 (15), 223 (45), 195 (11), 165 (100),

133 (12), 123 (13), 107 (5); Anal. Cald for C20H25N3O5S : C, 57.26; H, 6.01; N,

10.02; S, 7.64; Found: C, 57.21; H, 6.11; N, 10.12; S, 7.69.

1-[3-(4-Methoxyphenyl)propanoyl]-4-(2-methoxyphenyl)thiosemicarbazide (88o): Yield: 80%; m.p: 189-191oC; Rf: 0.34 (petroleum ether : acetone, 6:4); IR

(KBr, νmax, cm-1): 3383-3116 (NH Stretching), 1668 (C=O), 1520 (C=C), 1235

(C=S); 1H NMR (DMSO-d 6, δ ppm ): 9.99 (1H, s, NH), 9.67 (1H, s, NH), 9.53

(1H, s, NH), 7.43 (2H, d, J = 8.4 Hz, H-2′,6′), 7.29 (2H, d, J = 8.4 Hz, H-3′,5′),

6.84-6.33 (4H, m, H-3,4,5,6), 3.83 (3H, s, OCH3), 3.73 (3H, s, OCH3), 2.87(2H, t,

J = 7.1 Hz, CH2), 2.56 (2H, t, J = 7.5 Hz, CH2); 13C NMR (DMSO-d 6, δ ppm ):

Page 268: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

264

169.3 (C=O), [157.8, 157.4, 132.7, 129.6, 128.5, 126.3, 123.3,114.2] (Ar-C),

55.6-55.4 (OCH3), 35.7 (CH2), 29.9 (CH2); GCMS (DMF, m/z, %): 359 (4, M+),

325 (22), 237 (13), 194 (17), 178 (25), 167 (100), 163 (45), 135 (23), 123 (9), 107

(18); Anal. Cald for C18H21N3O3S : C, 60.15; H, 5.89; N, 11.69; S, 8.92; Found: C,

60.19; H, 5.75; N, 11.65; S, 8.91.

1-[2-(2,4-Dichlorophenoxy)propanoyl]-4-(2-methoxyphenyl)thiosemi-

carbazide (88p): Yield: 78%; m.p: 186-187°C; Rf: 0.37 (Petroleum ether :

acetone; 6:4); IR (KBr, νmax, cm-1): 3250-3100 (NH Stretching), 1658 (C=O),

1568 (C=C), 1247 (C=S); 1H-NMR (DMSO-d 6, δ ppm ): 9.98 (1H, s, NH), 9.55

(1H, s, NH), 9.38 (1H, s, NH), 7.63-7.15 (3H, m, H-3′,5′,6′), 4.79 (1H, q, CH),

6.80-6.39 (2H, m, H-3,4,5,6), 3.80 (3H, s, OCH3), 2.03 (3H, d, CH3); 13C NMR

(DMSO-d 6, δ ppm ): 167.4 (C=O), 157.2, 153.5, 129.8, 128.3), 125.4, 124.3,

122.8, 115.7](Ar-C), 74.25 (CH), 19.13 (CH3); GCMS (DMF, m/z, %): 414 (3, M+),

380 (21), 291(25), 248 (54), 232 (20), 216 (19), 189 (21), 167 (100), 133 (22),

123 (3), 107 (3); Anal. Cald for C17H17Cl2N3O3S : C, 49.28; H, 4.14; N, 10.14; S,

7. 74; Found: C, 49.18; H, 4.10; N, 10.17; S, 7. 70.

1-[4-(2,4-Dichlorophenoxy)butanoyl]-4-(2-methoxyphenyl)thiosemi- carbazide (88q): Yield: 85%; m.p: 155-156oC; Rf: 0.39(petroleum ether:

acetone, 6:4); IR (KBr, νmax, cm-1): 3311-3019 (NH Stretching), 1670 (C=O),

1605 (C=C); 1H NMR (DMSO-d 6, δ ppm ): 10.05 (1H, s, NH), 9.61(1H, s, NH),

9.02 (1H, s, NH), 7.88-6.89 (7H, m, Ar-H), 4.11 (2H, t, J = 6.3 Hz, CH2), 3.76

(3H, s, OCH3), 2.39 (2H, t, J = 7.2 Hz, CH2), 2.02 (2H, t, J = 7.2 Hz, CH2); 13C

NMR (DMSO-d 6, δ ppm ): 169.1 (C=O), [153.4, 151.6, 129.7, 128.5, 128.5,

126.4, 124.8, 122.8, 120.2, 115.5, 111.8] (Ar-C), 68.7 (CH2), 56.1 (OCH3), 30.0

(CH2), 24.7 (CH2); GCMS (DMF, m/z, %): 428(7, M+), 394 (11), 322 (21), 262

(15), 246 (33), 231(15), 203 (31), 167(100), 133 (2), 123 (13), 107 (8); 63 (5), 51

(7), 31 (2); Anal. Cald for C18H19Cl2N3O3S : C, 50.47; H, 4.47; N, 9.81; S, 07.49;

Found: C, 50.47; H, 4.45; N, 9.88; S, 7.55.

Page 269: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

265

1-[3-(4-Methoxyphenyl)propanoyl]-4-(4-methoxyphenyl)thiosemicarbazide (88r): Yield: 83%; m.p: 165-167oC; Rf: 0.35 (petroleum ether : acetone, 6:4); IR

(KBr, νmax, cm-1): 3432-3210 (NH Stretching), 1675 (C=O), 1605 (C=C), 1252

(C=S); 1H NMR (DMSO-d 6, δ ppm ): 9.87 (1H, s, NH), 9.47 (1H, s, NH), 9.33

(1H, s, NH), 7.23 (2H, d, J = 8.4Hz, H-2′,6′), 7.14 (2H, d, J = 8.4 Hz, H-3′,5′),

6.89 (2H, d, J = 8.4Hz, H-3,5), 6.83 (2H, d, J = 8.4 Hz, H-2,6), 3.74 (3H, s,

OCH3), 3.69 (3H, s, OCH3), 2.79 (2H, t, J = 7.5 Hz, CH2), 2.46 (2H, t, J = 7.5 Hz,

CH2); 13C NMR (DMSO-d 6, δ ppm ): 171.7 (C=O), [157.9, 157.2, 133.4, 132.3,

129.6, 114.2] (Ar-C), 55.6-55.4 (OCH3), 35.7 (CH2), 29.9 (CH2); GCMS (DMF,

m/z, %): 359 (19, M+), 325 (20), 237 (8), 194 (17), 178 (21), 167 (100), 163 (23),

135 (29), 123 (19), 107 (1); Anal. Cald for C18H21N3O3S : C, 60.15; H, 5.89; N,

11.69; S, 8.92; Found: C, 60.11; H, 5.79; N, 11.65; S, 8.99.

1-[2-(4-Bromophenoxy)acetyl]-4-(4-methoxyphenyl)thiosemicarbazide (88s): Yield: 80%; m.p: 141-143oC; Rf: 0.42 (petroleum ether : acetone, 6:4); IR (KBr,

νmax, cm-1): 3326-3100 (NH Stretching), 1663 (C=O), 1525 (C=C), 1254 (C=S);

1H-NMR (DMSO-d 6, δ ppm ): 10.24 (1H, s, NH), 9.56 (2H, s, NH), 7.47 (2H, d, J

= 8.7 Hz, H-3′,5′), 7.24 (2H, d, J = 8.7Hz, H-2′,6′), 6.98 (2H, d, J = 8.4Hz, H-3,5),

6.90 (2H, d, J = 8.7 Hz, H-2,6), 4.61 (2H, s, CH2), 3.76 (3H, s, OCH3); 13C NMR

(DMSO-d 6, δ ppm ): 171.7 (C=O), [157.5, 157.3, 132.5, 129.6, 128.9, 117.4,

113.7] (Ar-C), 66.4 (CH2), 55.6 (OCH3); GCMS (DMF, m/z, %): 410 (21, M+), 377

(14), 287 (20), 244 (13), 228 (19), 213 (45), 185 (11), 165 (100), 133 (12), 123

(13), 107 (15); Anal. Cald for C16H16BrN3O3S : C, 46.84; H, 3.93; N, 10.24; S,

7.82; Found: C, 46.89; H, 3.95; N, 10.27; S, 7.88.

1-[2-(2,4-Dichlorophenoxy)acetyl]-4-(4-methoxyphenyl)thiosemicarbazide (88t): Yield: 79%; m.p: 134-135oC; Rf: 0.35 (petroleum ether : acetone, 6:4); IR

(KBr, νmax, cm-1): 3142-3001 (NH Stretching), 1675 (C=O), 1605 (C=C), 1248

(C=S); 1H NMR (DMSO-d 6, δ ppm ): 10.20 (1H, s, NH), 9. 60 (2H, s, NH), 7.60-

6.89 (7H, m, Ar-H), 4.78 (2H, s, CH2), 3.74 (3H, s, OCH3); 13C NMR (DMSO-d 6,

Page 270: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

266

δ ppm ): 169.4 (C=O), [160.2, 157.3, 132.2, 129.8, 129.7, 128.4, 128.3, 116.1,

114.8] (Ar-C), 67.4 (CH2), 55.9 (OCH3); GCMS (DMF, m/z, %): 400 (11, M+), 365

(27), 277 (13), 234 (43), 218 (4), 203 (29), 175 (19), 165 (100), 133 (2), 123 (12),

107 (8); Anal. Cald for C16H15Cl2N3O3S : C, 48.01; H, 3.78; N, 10.50; S, 8.01;

Found: C, 48.07; H, 3.71; N, 10.40; S, 8.11.

1-(2,5-Difluorobenzoyl)-4-(4-methoxyphenyl)thiosemicarbazide (88u): Yield:

84%; m.p: 119-120oC; Rf: 0.37 (petroleum ether : acetone, 6:4); IR (KBr, νmax,

cm-1): 3254-3006 (NH Stretching), 1663 (C=O), 1509 (C=C), 1245 (C=S); 1H

NMR (DMSO-d 6, δ ppm ): 10.66 (1H, s, NH), 9.95 (1H, s, NH), 9.23 (1H, s, NH),

7.52-7.38 (3H, m, H-3′,4′,6′), 7.16 (2H, d, J = 7.5 Hz, H-3,5), 7.06 (2H, d, J = 7.8

Hz, H-2,6), 3.81 (3H, s, OCH3); 13C NMR (DMSO-d 6, δ ppm ): 166.3 (C=O),

[160.7, 153.6, 129.8, 126.2, 123.5, 120.2, 115.9] (Ar-C), 55.6 (OCH3); GCMS

(DMF, m/z, %): 337 (4, M+), 303 (19), 215 (22), 172 (15), 165 (100), 156 (5), 141

(54), 133 (14), 123 (19), 113 (8), 107 (21); Anal. Cald for C15H13F2N3O2S : C,

53.41; H, 3.88; N, 12.46; S, 9.91; Found: C, 53.46; H, 3.77; N, 12.42; S, 9.96.

1-[3-(3,4,5-Trimethoxyphenyl)propanoyl]-4-(4-methoxyphenyl)thiosemi- carbazide (88v): Yield: 85%; m.p: 164-165oC; Rf: 0.39 (petroleum ether :

acetone, 6:4); IR (KBr, νmax, cm-1): 3330-3016 (NH Stretching), 1645 (C=O),

1555 (C=C), 1236 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 11.03 (1H, s, NH), 10.45

(1H, s, NH), 9.55 (1H, s, NH), 7.40 (1H, dd, J = 7.8 Hz, H-2, 6), 7.23 (1H, d, J =

8.1Hz, H-3,5), 7.15 (2H, s, H-2’,6’), 2.41 (2H, t, J = 6.9 Hz, CH2 ), 2.31 (2H, t, J =

6.9 Hz, CH2); 13C NMR (DMSO-d 6, δ ppm ): 187.8 (C=S), 167.5 (C=O), [161.0,

151.1, 150.7, 138.1, 136.2, 133.8, 130.1, 118.8, 110.2, 109.7, 105.1] (Ar-C),

55.8-56.0 (OCH3), 41.4 (CH2), 39.1 (CH2); GCMS (DMF, m/z, %): 419(17, M+),

385 (12), 254 (25), 238 (10), 223 (45), 195 (11), 165 (100), 133 (12), 123 (13),

107 (5); Anal. Cald for C20H25N3O5S : C, 57.26; H, 6.01; N, 10.02; S, 7.64;

Found: C, 57.19; H, 6.11; N, 10.22; S, 7.69.

Page 271: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

267

1-(3,4-Dimethoxybenzoyl)-4-(4-methoxyphenyl)thiosemicarbazide (88w): Yield: 80%; m.p: 148-149oC; Rf: 0.40 (petroleum ether : acetone, 6:4); IR (KBr,

νmax, cm-1): 3400-3180 (NH Stretching), 1668 (C=O), 1596 (C=C), 1262 (C=S);

1H NMR (DMSO-d 6, δ ppm ): 10.37 (1H, s, NH), 9.68 (1H, s, NH), 9.57 (1H, s,

NH), 7.59 (1H, dd, J = 8.4, 1.8 Hz, H-6 ), 7.54 (1H, d, J = 1.8 Hz, H-2), 7.31 (2H,

d, J = 9.0 Hz, H-3’, 5’), 7.05 (1H, d, J = 8.4 Hz, H-5), 6.92 (2H, d, J = 9.0 Hz, H-

2’,6’), 3.88 (6H, s, OCH3), 3.82 (3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ):

166.0 (C=O), [157.1, 152.1, 127.9, 121.8, 113.6] (Ar-C), 55.8-56.0 (OCH3);

GCMS (DMF, m/z, %): 361(12, M+), 327 (32), 239 (27), 196 (19), 180 (26), 165

(100), 165 (21), 137 (12), 123 (13), 107 (8); Anal. Cald for C17H19N3O4S : C,

56.50; H, 5.30; N, 11.63; S, 8.87; Found: C, 56.57; H, 5.33 N, 11.69; S, 8.77.

7.8 General procedure for the synthesis of 1,2,4 Triazole-3-thiones

The respective thiosemicarbazide (1.4 mmol) was refluxed in 25mL of 4N

aqueous sodium hydroxide solution. The reaction was monitored by TLC. After

completion of reaction, the reaction mixture was cooled to room temperature and

filtered. The filtrate was neutralized with 6N hydrochloroic acid to precipitate the

triazole which was filtered and recrystalized from aqueous ethanol.

5-[1-(2,4-Dichlorophenoxy)methyl]-4-(cyclohexyl)-4H-1,2,4-triazole-3-thione (89a): Yield: 58%; m.p.: 178-180°C; Rf : 0.36 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3409 (NH stretching), 3038-2873 (CH stretching), 1509 (C=N),

1591-1484 (C=C), 1271 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 12.03 (1H, s, NH),

7.59 (1H, d, J = 2.1Hz, H-3′), 7.12 (1H, d, J = 7.8 Hz, H-5′), 6.82 (1H, d, J = 8.1

Hz, H-6′), 4.69 (2H, s, CH2), 3.23 (1H, s, H-1), 1.75-1.13 (10H, m, cyclohexyl H);

13C NMR (DMSO-d 6, δ ppm): 169.5 (C=S), 161.2 (C=N), [153.4, 152.3, 134.1,

130.8, 129.9] (Ar-C), 77.8 (CH2), [53.1, 32.4, 30.6, 25.2] (Cycylohexyl-C); GCMS

(DMF, m/z, %): 358 (10, M+), 357 (2), 325 (17), 298 (21), 216 (14), 215 (2), 201

Page 272: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

268

(100), 175 (13), 141(8); Anal. Cald for C15H17Cl2N3OS : C, 50.28; H, 4.78; N,

11.73; S, 8.95; Found: C, 50.26; H, 4.70; N, 11.74; S, 8.90.

5-(3,4,5-Trimethoxyphenylethyl)-4-(cyclohexyl)-4H-1,2,4-triazole-3-thione (89b): Yield: 61%; m.p.: 234-235°C; Rf : 0.31 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3101 (NH stretching), 3038-2873 (CH stretching), 1501(C=N),

1601-1384 (C=C), 1276 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 13.04 (1H, s, NH),

6.33 (2H, s, H-2′,6′), 4.01 (1H, s, H-1), 1.71-1.07 (10H, m, cyclohexyl-H), 2.72-

2.54 (4H, m, 2 × CH2); 13C NMR (DMSO-d 6, δ ppm ): 169.5 (C=S), 152.4 (C=N),

[153.8, 135.1, 132.8, 105.9] (Ar-C), 60.3-56.1 (OCH3), [53.1, 32.4, 30.6, 27.6,

25.2](Cycylohexyl-C & CH2); GCMS (DMF, m/z, %): 377 (3, M+), 376 (10), 344

(23), 236 (12), 221 (100), 195 (3), 141(5), 83 (54); Anal. Cald for C19H27N3O3S :

C, 60.45; H, 7.21; N, 11.13; S, 0.49; Found: C, 60.48; H, 7.24; N, 11.17; S, 8.55.

5-(3,5-Difluorophenyl)-4-(cyclohexyl)-4H-1,2,4-triazole-3-thione (89c): Yield:

67%; m.p.: 221-223°C; Rf : 0.25 (Petroleum ether : acetone; 6:4); IR (KBr, νmax,

cm-1): 3411 (NH stretching), 3018-2853 (CH stretching), 1516 (C=N), 1589-1481

(C=C), 1255 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 12.95 (1H, s, NH), 7.59 (2H,

ddd, J = 12.5, 2.4 Hz, H-2′,6′), 7.05 (1H, m, H-4′), 4.06 (1H, s, H-1), 1.70-1.03

(10H, m, cyclohexyl H); 13C NMR (DMSO-d 6, δ ppm ): 175.0 (C=S), 154.7

(C=N), [168.2, 132.8, 107.5, 106.7] (Ar-C), [56.3, 32.3, 29.6, 24.3] (Cycylohexyl-

C); GCMS (DMF, m/z, %): 295 (9, M+), 262 (17), 236 (12), 154 (13), 141 (100),

139 (12), 113 (50); Anal. Cald for C14H15F2N3S : C, 56.93; H, 5.12; N, 14.23;

S,10.86; Found: C, 56.85; H, 5.17; N, 14.28; S, 10.81.

5-(3,5-Dimethoxyphenyl)-4-(cyclohexyl)-4H-1,2,4-triazole-3-thione (89d): Yield: 51%; m.p.: 120-122°C; Rf : 0.36 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3331 (NH stretching), 3090-2893 (CH stretching), 1521(C=N), 1571-

1454 (C=C), 1266 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.65 (1H, s, NH), 6.45

(1H, dd, J = 2.4 Hz, H-4′), 6.37 (2H, d, J = 2.1 Hz, H-2′,6′), 3.95 (1H, s, H-1),

Page 273: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

269

3.73 (6H, s, OCH3), 1.66-1.13 (10H, m, cyclohexyl H); 13C NMR (DMSO-d 6, δ

ppm ): 173.2 (C=S), 152.0 (C=N), [162.6, 131.8, 104.5, 102.7](Ar-C), 56.2-55.4

(OCH3), [57.9, 33.3, 27.6, 25.3] ( Cycylohexyl-C); GCMS (DMF, m/z, %): 319 (29,

M+), 286 (21), 260 (7), 176 (5), 164 (21), 141 (100), 137 (14); Anal. Cald for

C16H21N3O2S: C, 60.16; H, 6.63; N, 13.16; S, 10.04; Found: C, 60.12; H, 6.69; N,

13.14; S, 10.17.

5-(3,4,5-Trimethoxyphenylethyl)-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89e): Yield: 47%; m.p.: 218-219°C; Rf : 0.31 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3301 (NH stretching), 3038-2873 (CH stretching), 1611

(C=N), 1601-1455 (C=C), 1256 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.67 (1H,

s, NH), 7.53 (1H, ddd, J = 7.8, 1.8 Hz, H-5), 7.26 (1H, dd, J = 7.8, 0.6 Hz, H-3),

7.18 (1H, dd, J = 7.8, 1.8 Hz, H-6), 7.09 (1H, ddd, J = 7.5, 0.9 Hz, H-4), 6.29 (2H,

s, H-2′,6′), 3.76-3.59 (12H, s, OCH3), 2.72-2.54 (4H, m, 2 × CH2); 13C NMR

(DMSO-d 6, δ ppm ): 168.3 (C=S), 152.4 (C=N), [154.9, 153.1, 136.2, 131.8,

130.6, 122.2, 121.2, 113.3, 105.9] (Ar-C), 60.3-56.1 (OCH3), 32.4 (CH2), 27.6

(CH2); GCMS (DMF, m/z, %): 401 (33, M+), 386 (10), 368 (22), 287 (9), 206 (3),

181 (100), 197 (6), 148 (13), 137 (7), 120 (4), 105(3), 92 (3),77 (11), 65 (5), 51

(4), 39 (2); Anal. Cald for C20H23N3O4S : C, 59.83; H, 5.77; N, 10.47; S, 7.99;

Found: C, 59.89; H, 5.71; N, 10.43; S, 8.05.

5-(3,5-Dimethoxyphenyl)-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89f): Yield: 49%; m.p.: 197-198°C; Rf : 0.35 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3291 (NH stretching), 3108-2973 (CH stretching), 1501 (C=N),

1581-1474 (C=C), 1241 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 14.07 (1H, s, NH),

7.49 (1H, ddd, J = 9.0,1.5 Hz, H-5), 7.39 (1H, dd, J = 7.8, 1.5 Hz, H-3), 7.17(1H,

dd, J = 7.8, 0.6 Hz, H-6), 7.09 (1H, ddd, J = 7.5, 1.2 Hz, H-4), 6.52 (1H, dd, J =

4.5, 2.4 Hz, H-4′), 6.45 (2H, d, J = 2.1 Hz, H-2′,6′), 3.58 (9H, s, OCH3); 13C NMR

(DMSO-d6, δ ppm ): 169.4 (C=S), 151.0 (C=N), [160.6, 155.0, 130.8, 128.0,

123.6, 121.4, 113.3, 105.5, 102.7] (Ar-C), 56.3-55.6 (OCH3); GCMS (DMF, m/z,

%): 343 (45, M+), 310 (100), 280 (2), 254 (3), 163 (8), 149 (15), 137(3), 120 (11),

Page 274: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

270

105(7), 92 (6),77 (17), 65 (5), 51(4), 39 (2); Anal. Cald for C17H17N3O3S : C,

59.46; H, 4.99; N, 12.24; S, 9.34; Found: C, 59.49; H, 4.91; N, 12.28; S, 9.36.

5-(2,5-Difluorophenyl)-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89g): Yield: 75%; m.p.: 133-134°C; Rf : 0.34 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3361 (NH stretching), 3056-2943 (CH stretching), 1531 (C=N), 1584-

1484 (C=C), 1244 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.46 (1H, s, NH), 7.61

(1H, dd, J = 12.5,2.1 Hz, H-2′), 7.49 (1H, ddd, J = 9.0,1.5 Hz, H-5), 7.39 (1H, dd,

J = 7.8, 1.5 Hz, H-3), 7.21 (2H, m, H-4′,5′), 7.17 (1H, dd, J = 7.8, 0.6 Hz, H-6),

7.09 (1H, ddd, J = 7.5, 1.2 Hz, H-4), 3.81 (3H, s, OCH3); 13C NMR (DMSO-d6, δ

ppm ): 174.0 (C=S), 153.6 (C=N), [162.2, 159.7, 130.8, 128.3, 122.6, 121.4,

118.2 113.5, 111.5] (Ar-C), 56.3 (OCH3); GCMS (DMF, m/z, %): 319 (9, M+), 286

(15), 260 (51), 180 (22), 166 (41), 153 (32), 139 (100), 113 (8), 107 (5); Anal.

Cald for C15H11F2N3OS : C, 56.42; H, 3.47; N, 13.16; S, 10.04; Found: C, 56.48;

H, 3.45; N, 13.10; S, 10.14.

5-[1-(2,4-Dichlorophenoxy)methyl]-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89h): Yield: 58%; m.p.: 201-202°C; Rf : 0.40 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3419 (NH stretching), 3081-2813 (CH stretching), 1498

(C=N), 1586-1456 (C=C), 1286 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.03 (1H,

s, NH), 7.66 (1H, s, H-3′), 7.49 (1H, d, J = 7.8 Hz, H-5′), 7.21 (1H, d, J = 8.1Hz,

H-6′), 7.19 (1H, ddd, J = 9.0,1.5 Hz, H-5), 7.13 (1H, dd, J = 7.8, 1.5 Hz, H-3),

7.07 (1H, dd, J = 7.8, 0.6 Hz, H-6), 6.89 (1H, ddd, J = 7.5, 1.2 Hz, H-4), 4.69

(2H, s, CH2), 3.85 (3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ): 165.0 (C=S),

152.3 (C=N), [160.4, 155.5, 143.1, 131.8, 130.9, 128.6, 124.6, 123.4, 122.4,

118.8, 115.5] (Ar-C), 79.2 (CH2), 56.8 (OCH3); GCMS (DMF, m/z, %): 382 (33,

M+), 348 (10), 221 (22), 216 (25), 203 (100), 175 (13), 166 (8), 107 (15); Anal.

Cald for C16H13Cl2N3O2S : C, 50.27; H, 3.43; N, 10.99; S, 8.39; Found: C, 50.24;

H, 3.49; N, 10.98; S, 8.49.

Page 275: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

271

5-(3,5-Difluorophenyl)-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89i): Yield: 61%; m.p.: 169-170°C; Rf : 0.38 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3300 (NH stretching), 3015-2903 (CH stretching), 1507 (C=N), 1588-

1414 (C=C), 1244 (C=S); 1H NMR (DMSO-d6, δ ppm ): 14.06 (1H, s, NH), 7.52

(2H, dd, J = 15.5, 2.8 Hz, H-2′,6′), 7.43 (1H, m, H-4′), 7.39 (1H, ddd, J = 8.4,1.5

Hz, H-5), 7.27 (1H, dd, J = 7.5, 1.8 Hz, H-3), 7.13 (1H, dd, J = 7.8, 1.5 Hz, H-6),

7.09 (1H, ddd, J = 7.5, 1.2 Hz, H-4), 3.88 (3H, s, OCH3); 13C NMR (DMSO-d6, δ

ppm ): 172.8 (C=S), 155.6 (C=N), [164.2, 159.0,133.0, 128.8, 123.2, 121.4,

113.5, 108.5, 106.3] (Ar-C), 55.1 (OCH3); GCMS (DMF, m/z, %): 319 (18, M+),

286 (9), 260 (42), 180 (20), 166 (68), 153 (21), 139 (100), 113 (13), 107 (9); Anal. Cald for C15H11F2N3OS : C, 56.42; H, 3.47; N, 13.16; S, 10.04; Found: C,

56.48; H, 3.42; N, 13.14; S, 10.13.

5-[1-(4-Bromophenoxy)methyl]-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89j): Yield:67%; m.p.: 154-155°C; Rf : 0.36 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3333 (NH stretching), 3101-2804 (CH stretching), 1513

(C=N), 1594-1384 (C=C), 1271 (C=S); 1H NMR (DMSO-d6, δ ppm ): 2.74 (1H, s,

NH), 7.71 (1H, dd, J = 8.4,1.5 Hz, H-3), 7.55 (2H, m, H-4,5), 7.19 (1H, d, J = 7.5

Hz, H-6), 6.90 (2H, d, J = 8.1 Hz, H-2′,6′), 6.73 (2H, d, J = 7.8 Hz, H-3′,5′), 4.71

(2H, s, CH2), 3.85 (3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ): 170.5 (C=S),

154.0 (C=N), [163.1, 158.9, 134.1, 133.9, 129.6, 127.6, 123.5, 118.3, 115.2] (Ar-

C), 81.2 (CH2), 56.0 (OCH3); GCMS (DMF, m/z, %): 392 (11, M+), 358 (10), 231

(12), 226 (23), 213 (100), 185 (19), 166 (8), 107 (15); Anal. Cald for

C16H14BrN3O2S : C, 48.99; H, 3.60; N, 10.71; S, 8.17; Found: C, 48.90; H, 3.69;

N, 10.73; S, 8.12.

5-[2-(4-Methoxyphenyl)ethyl]-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89k): Yield: 51%; m.p.: 204-205°C; Rf : 0.31 (Petroleum ether: acetone; 6:4); IR

(KBr, νmax, cm-1): 3291 (NH stretching), 3104-2903 (CH stretching), 1521 (C=N),

1571-1464 (C=C), 1265 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.42 (1H, s, NH),

7.52 (1H, dd, J = 8.4,1.5 Hz, H-4), 7.25 (2H, m, H-4,5), 7.10 (1H,d, J = 7.5 Hz, H-

Page 276: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

272

6), 6.95 (2H, d, J = 8.1 Hz, H-2′,6′), 6.78 (2H, d, J = 7.8 Hz, H-3′,5′), 3.75 (3H, s,

OCH3), 3.68 (3H, s, OCH3), 2.71 (2H, t, J = 7.5 Hz, CH2), 2.53 (2H, t, J = 7.5 Hz,

CH2); 13C NMR (DMSO-d6, δ ppm ): 168.3 (C=S), 152.4 (C=N), [158.1, 154.9,

132.3, 131.8, 130.6, 129.6, 122.2, 121.9, 114.2, 113.2, 56.3-55.4 (OCH3), 30.8

(CH2), 27.6 (CH2); GCMS (DMF, m/z, %): 341(3, M+), 324 (17), 236 (5), 227

(43),188 (4), 134 (3), 121 (100), 105 (3), 92 (13),77 (21), 65 (7), 51(6), 39 (2);

Anal. Cald for C18H19N3O2S : C, 63.32; H, 5.61; N, 12.31; S, 9.39; Found: C,

63.36; H, 5.62; N, 12.39; S, 9.31.

5-[1-(2,4-Dichlorophenoxy)ethyl]-4-(2-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89l): Yield: 54%; m.p.: 165-166°C; Rf : 0.35 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3341 (NH stretching), 3054-2813 (CH stretching), 1491

(C=N), 1601-1484 (C=C), 1279 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 12.93 (1H,

s, NH), 7.62 (1H, s, H-3′), 7.59 (1H, d, J = 7.5 Hz, H-5′), 7.48 (1H, d, J = 8.4 Hz,

H-6′), 7.31 (1H, ddd, J = 9.0,1.5 Hz, H-5), 7.23 (1H, dd, J = 7.5, 1.5 Hz, H-3),

7.17 (1H, dd, J = 7.8, 0.6 Hz, H-6), 6.99 (1H, ddd, J = 7.5, 1.2 Hz, H-4), 4.41

(1H, s, CH2), 3.70 (3H, s, OCH3), 1.70 (3H, s, CH2); 13C NMR (DMSO-d 6, δ ppm

): 168.7 (C=S), 156.6 (C=N), [159.4, 151.5, 132.8, 129.9, 129.6, 129.6, 126.6,

124.4, 123.4, 116.8, 114.5] (Ar-C), 72.2 (CH2), 55.5 (OCH3), 15.2 (CH2); GCMS

(DMF, m/z, %): 396 (7, M+), 362 (21), 230 (9), 215 (16), 216 (100), 189 (32), 180

(13), 166 (3), 107 (15); Anal. Cald for C17H15Cl2N3O2S : C, 51.52; H, 3.82; N,

10.60; S, 8.09; Found: C, 51.58; H, 3.85; N, 10.67; S, 8.13.

5-(3,4,5-Trimethoxyphenylethyl)-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89m): Yield: 56%; m.p.: 169-170°C; Rf : 0.34 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3241 (NH stretching), 3038-2933 (CH stretching), 1481

(C=N), 1551-1481 (C=C), 1257 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.21(1H, s,

NH), 7.32 (1H, d, J = 7.8 Hz, H-3,5), 7.08 (1H, d, J = 7.5 Hz, H-2,6), 6.79 (1H, d,

J = 1.8 Hz, H-2′,6′), 3.84-3.61 (12H, s, OCH3), 2.73-2.50 (4H, m, CH2); 13C NMR

(DMSO-d 6, δ ppm ): 169.1 (C=S), 153.2 (C=N), [155.9, 151.1, 138.2, 134.8,

Page 277: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

273

128.6, 126.2, 115.3, 105.9] (Ar-C), 58.3-56.1 (OCH3), 32.4 (CH2), 30.6 (CH2); GCMS (DMF, m/z, %): 401 (25, M+), 386 (19), 368 (20), 287 (19), 206 (23), 181

(100), 197 (66), 148 (33), 137 (37), 120 (14), 105 (13), 92 (13), 77 (18), 65 (5),

51(4), 39 (2); Anal. Cald for C20H23N3O4S : C, 59.83; H, 5.77; N, 10.47; S, 7.99;

Found: C, 59.80; H, 5.76; N, 10.35; S, 8.15.

5-(3,5-Dimethoxyphenyl)-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89n): Yield: 75%; m.p.: 153-154°C; Rf : 0.39 (Petroleum ether : acetone; 6:4);

IR (KBr, νmax, cm-1): 3189 (NH stretching), 3031-2873 (CH stretching), 1497

(C=N), 1571-1464 (C=C), 1275 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.37 (1H,

s, NH), 7.53 (1H, d, J = 9.0 Hz, H-3,5), 7.24 (1H, d, J = 7.5 Hz, H-2,6), 6.72 (1H,

dd, J = 4.5, 2.4 Hz, H-4′), 6.51 (2H, d, J = 2.1 Hz, H-2′,6′), 3.71-3.61 (9H, s,

OCH3); 13C NMR (DMSO-d6, δ ppm ): 168.7 (C=S), 151.4 (C=N), [163.4, 157.2,

132.8, 128.0, 127.6, 117.3, 108.5, 104.7] (Ar-C), 56.3-55.0 (OCH3); GCMS

(DMF, m/z, %): 343 (39, M+), 310 (100), 280 (12), 254 (13), 163 (18), 149 (25),

137 (30), 120 (10), 105(12), 92 (16),77 (11), 65 (15), 51(6), 39 (8); Anal. Cald for

C17H17N3O3S : C, 59.46; H, 4.99; N, 12.24; S, 9.34. Found: C, 59.46; H, 4.94; N,

12.25; S, 9.30.

5-(2,5-Difluorophenyl)-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89o): Yield: 58%; m.p.: 173-175°C; Rf : 0.36 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3181 (NH stretching), 3001-2874 (CH stretching), 1502 (C=N), 1601-

1414 (C=C), 1245 (C=S); 1H NMR (DMSO-d6, δ ppm ): 2.09 (1H, s, NH), 7.54

(1H, dd, J = 13.0, 2.4 Hz, H-2′), 7.42 (1H, ddd, J = 11.3,1.0 Hz, H-3,5), 7.29 (2H,

m, H-4′,5′), 7.07 (1H, dd, J = 7.8, 0.6 Hz, H-2,6), 3.88 (3H, s, OCH3); 13C NMR

(DMSO-d6, δ ppm ): 175.1 (C=S), 154.38 (C=N), [160.5, 157.7, 129.3, 127.6,

121.4, 118.2, 116.5, 114.5, 113.5] (Ar-C), 56.7 (OCH3); GCMS (DMF, m/z, %): 319 (17, M+), 286 (20), 260 (35), 180 (29), 166 (13), 153 (32), 139 (100), 113

(18), 107 (15); Anal. Cald for C15H11F2N3OS : C, 56.42; H, 3.47; N, 13.16; S,

10.04; Found: C, 56.45; H, 3.49; N, 13.15; S, 10.16.

Page 278: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

274

5-[1-(2,4-Dichlorophenoxy)methyl]-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89p): Yield: 42%; m.p.: 222-223°C; Rf : 0.31 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3341 (NH stretching), 3018-2813 (CH stretching), 1514

(C=N), 1587-1411 (C=C), 1255 (C=S); 1H NMR (DMSO-d6, δ ppm ): 12.73 (1H,

s, NH), 7.51 (1H, s, H-3′), 7.44 (1H, d, J = 7.8 Hz, H-5′), 7.29 (1H, d, J = 8.1 Hz,

H-6′), 7.14 (1H, d, J = 9.0, 1.5 Hz, H-3,5), 7.10 (1H, d, J = 7.8, 1.5 Hz, H-2,6),

4.55 (2H, s, CH2), 3.74 (3H, s, OCH3); 13C NMR (DMSO-d 6, δ ppm ): 167.0

(C=S), 153.3 (C=N), [156.4, 154.5, 132.4, 131.8, 130.9, 128.6, 126.6, 123.4,

118.8, 115.5] (Ar-C), 77.2 (CH2), 56.8 (OCH3); GCMS (DMF, m/z, %): 382 (29,

M+), 348 (19), 221 (32), 216 (20), 203 (100), 175 (33), 166 (81), 107 (19); Anal.

Cald for C16H13Cl2N3O2S : C, 50.27; H, 3.43; N, 10.99; S, 8.39; Found: C, 50.24;

H, 3.41; N, 10.95; S, 8.35.

5-(3,5-Difluorophenyl)-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89q): Yield: 66%; m.p.: 229-230°C; Rf : 0.35 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3349 (NH stretching), 3008-2803 (CH stretching), 1481 (C=N), 1571-

1454 (C=C), 1248 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.36 (1H, s, NH), 7.61

(2H, dd, J = 15.5, 2.8 Hz, H-2′,6′), 7.47 (1H, m, H-4′), 7.31 (1H, ddd, J = 8.4,1.5

Hz, H-3,5), 7.07 (1H, dd, J = 7.8, 1.5 Hz, H-2,6), 3.81 (3H, s, OCH3); 13C NMR

(DMSO-d6, δ ppm ): 170.6 (C=S), 154.6 (C=N), [166.2, 157.0, 133.7, 128.8,

125.4, 115.5, 107.5, 106.3] (Ar-C), 55.9 (OCH3); GCMS (DMF, m/z, %): 319 (20,

M+), 286 (26), 260 (21), 180 (59), 166 (19), 153 (38), 139 (100), 113 (10), 107

(5); Anal. Cald for C15H11F2N3OS : C, 56.42; H, 3.47; N, 13.16; S, 10.04; Found:

C, 56.42; H, 3.46; N, 13.19; S, 10.03.

5-[1-(4-Bromophenoxy)methyl]-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89r): Yield: 75%; m.p.: 156-158°C; Rf : 0.34 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3425 (NH stretching), 3108-2913 (CH stretching), 1521

(C=N), 1551-1435 (C=C), 1247 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 2.51 (1H, s,

NH), 7.68 (2H, d, J = 8.4 Hz, H-3,5), 7.53 (2H, d, J = 7.8 Hz, H-3′,5′), 6.23 (2H,

d, J = 8.4 Hz, H-2′,6′), 6.81 (2H, d, J = 7.8 Hz, H-2,6), 4.44 (2H, s, CH2), 3.74

Page 279: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

275

(3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ): 169.3 (C=S), 153.2 (C=N), [160.1,

157.4, 135.1, 128.4, 127.6, 117.3, 115.2] (Ar-C), 78.2 (CH2), 56.43 (OCH3); GCMS (DMF, m/z, %): 392 (21, M+), 358 (61), 231 (32), 226 (12), 213 (100), 185

(12), 166 (28), 107 (9); Anal. Cald for C16H14BrN3O2S : C, 48.99; H, 3.60; N,

10.71; S, 8.17; Found: C, 48.97; H, 3.65; N, 10.63; S, 8.17.

5-[1-(4-Methoxyphenyl)ethyl]-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89s): Yield: 58%; m.p.: 102-103°C; Rf : 0.29 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3201(NH stretching), 3010-2870 (CH stretching), 1519 (C=N),

1601-1489 (C=C), 1276 (C=S); 1H NMR (DMSO-d6, δ ppm ): 13.15 (1H, s, NH),

7.59 (1H, d, J = 8.4 Hz, H-2′,6′), 7.35 (2H, d, J = 7.8 Hz, H-3′,5′), 6.95 (2H, d, J =

8.1 Hz, H-3,5), 6.73 (2H, d, J = 7.8 Hz, H-2,6), 3.70-3.62 (6H, s, OCH3), 3.02

(2H, t, J = 7.5 Hz, CH2), 2.14 (2H, t, J = 7.5 Hz, CH2); 13C NMR (DMSO-d6, δ

ppm ): 167.8 (C=S), 155.4 (C=N), [157.6, 135.3, 130.1,128.6, 127.2, 117. 9, 117.

2, 115.2] (Ar-C), 56.8-55.3 (OCH3), 35.8 (CH2), 20.6 (CH2); GCMS (DMF, m/z,

%): 341(17, M+), 324 (20), 236 (15), 227 (32),188 (24), 134 (32), 121 (100), 105

(23), 92 (23),77 (29), 65 (71), 51 (46), 39 (12); Anal. Cald for C18H19N3O2S : C,

63.32; H, 5.61; N, 12.31; S, 9.39; Found: C, 63.39; H, 5.62; N, 12.35; S, 9.36.

5-[1-(2,4-Dichlorophenoxy)ethyl]-4-(4-methoxyphenyl)-4H-1,2,4-triazole-3-thione (89t): Yield: 42%; m.p.: 208-209°C; Rf : 0.28 (Petroleum ether : acetone;

6:4); IR (KBr, νmax, cm-1): 3201 (NH stretching), 3038-2873 (CH stretching), 1505

(C=N), 1581-1444 (C=C), 1245 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 13.13 (1H,

s, NH), 7.58 (1H, s, H-3′), 7.49 (1H, d, J = 7.5 Hz, H-5′), 7.37 (1H, d, J = 8.4 Hz,

H-6′), 7.03 (1H, d, J = 7.8 Hz, H-3,5), 6.87 (1H, d, J = 7.5 Hz, H-2,6), 4.52 (2H,

s, CH2), 3.81 (3H, s, OCH3), 1.79 (2H, s, CH2); 13C NMR (DMSO-d6, δ ppm ):

169.5 (C=S), 154.6 (C=N), [158.4, 152.5, 132.5, 128.9, 128.6, 127.9, 126.6,

125.4, 124.4, 118.8, 115.5] (Ar-C), 72.2 (CH2), 55.5 (OCH3), 15.2 (CH2); GCMS

(DMF, m/z, %): 396 (14, M+), 362 (19), 230 (29), 215 (36), 216 (100), 189 (32),

180 (13), 166 (23), 107 (3);Anal. Cald for C17H15Cl2N3O2S : C, 51.52; H, 3.82; N,

10.60; S, 8.09. Found: C, 51.54; H, 3.80; N, 10.67; S, 8.11.

Page 280: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

276

7.9 General procedure for the synthesis of 1,3,4- Thiadiazoles

Each thiosemicarbazide (0.1 mmol) was added in portion to conc. sulphuric

acid (15 mL, 98%) at 0°C. The reaction mixture was heated at 120°C in an oil

bath for 10 minutes. The mixture was then stirred for 1 h at room temperature.

The reaction mixture was then poured into crushed ice and followed by dropwise

addition of aqueous ammonia (33%) till the appearance of a solid. The crude

1,3,4-thiadiazole was filtered, washed with water and recrystallized from acetic

acid-water (1:10).

Cyclohexylamino-5-(3,4,5-trimethoxyphenethyl)-1,3,4-thiadiazole (90a): Yield: 49%; m.p.: 134-135°C; Rf : 0.34 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3101 (NH stretching), 3038-2873 (CH stretching), 1501 (C=N), 1601-

1384 (C=C), 1276 (C=S); 1H NMR (DMSO-d 6, δ ppm ): 9.04 (1H, s, NH), 6.41

(2H, d, J = 2.4 Hz, H-2′,6′), 3.57 (1H, s, H-1), 1.87-1.17 (10H, m, cyclohexyl H),

2.92-2.74 (4H, m, 2 × CH2); 13C NMR (DMSO-d 6, δ ppm ): [173.5, 170.2] (C=N),

[153.5, 141.4, 134.8, 105.9] (Ar-C), 60.1-56.6 (OCH3), [55.2,34.2, 32.4, 29.6,

27.6, 22.2] (cyclohexyl-C & CH2 ); GCMS (DMF, m/z, %): 377 (19, M+), 295

(100), 279 (22), 221 (13), 156 (8), 98 (25), 58 (7); Anal. Cald for C19H27N3O3S :

C, 60.45; H, 7.21; N, 11.13; S, 8.49; Found: C, 60.42; H, 7.27; N, 11.13; S, 8.50.

Cyclohexylamino-5-(3,5-dimethoxyphenyl)-1,3,4-thiadiazole (90b): Yield:

51%; m.p.: 198-199°C; Rf : 0.38 (Petroleum ether : acetone; 6:4); IR (KBr, νmax,

cm-1): 3309 (NH stretching) , 3021-2881 (CH stretching), 1511 (C=N), 1611-

1385 (C=C); 1H NMR (DMSO-d6, δ ppm ): 10.05 (1H, s, NH), 6.39 (1H, dd, J =

3.2, 2.1 Hz, H-4′), 6.27 (2H, d, J = 2.1 Hz, H-2′,6′), 3.17 (1H, s, H-1), 3.88 (6H, s,

OCH3), 1.83-1.13 (10H, m, cyclohexyl H); 13C NMR (DMSO-d6, δ ppm ): [176.2,

167.2] (C=N), [163.3, 140.8, 107.5, 99.7] (Ar-C), 55.4 (OCH3), [54.9, 35.3, 27.1,

Page 281: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

277

25.3] (cyclohexyl-C); GCMS (DMF, m/z, %): 319 (7, M+), 237 (100), 221 (13),

177 (21), 163 (23), 141 (8), 98 (25), 83 (6), 74 (79), 58 (7); Anal. Cald for

C16H21N3O2S : C, 60.16; H, 6.63; N, 13.16; S, 10.04; Found: C, 60.14; H, 6.63; N,

13.15; S, 10.15.

2-Methoxyphenylamino-5-(3,4,5-trimethoxyphenethyl)-1,3,4-thiadiazole (90c): Yield: 48%; m.p.: 147-148°C; Rf : 0.36 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3291 (NH stretching), 3008-2821 (CH stretching), 1515 (C=N),

1591-1383 (C=C); 1H NMR (DMSO-d 6, δ ppm ): 9.87 (1H, s, NH), 7.57 (1H, ddd,

J = 7.8, 2.4Hz, H-5), 7.36 (1H, dd, J = 7.5, 1.8 Hz, H-3), 7.15 (1H, dd, J = 7.8, 1.8

Hz, H-6), 7.03 (1H, ddd, J = 7.5, 0.9 Hz, H-4), 6.59 (2H, s, H-2′,6′), 3.88-3.87

(12H, s, OCH3), 2.79-2.57 (4H, m, 2 × CH2); 13C NMR (DMSO-d6, δ ppm ):

[170.3, 156.5] (C=N), [153.1, 150.9, 138.2, 135.8, 132.2, 123.6, 120.2, 116.3,

107.9] (Ar-C), 59.3-56.5 (OCH3), 36.4 (CH2), 33.6 (CH2); GCMS (DMF, m/z, %):

401(33, M+), 295 (100), 279 (22), 235 (13), 221 (13), 165 (25), 156 (8), 107 (25),

58 (7); Anal. Cald for C20H23N3O4S : C, 59.83; H, 5.77; N, 10.47; S, 7.99; Found:

C, 59.85; H, 5.75; N, 10.47; S, 7.95.

2-Methoxyphenylamino-5-(3,5-dimethoxyphenyl)-1,3,4-thiadiazole (90d): Yield: 58%; m.p.: 189-191°C; Rf : 0.39(Petroleum ether: acetone; 6:4); IR (KBr,

νmax, cm-1): 3241 (NH stretching), 3018-2913 (CH stretching), 1521 (C=N), 1601-

1414 (C=C); 1H NMR (DMSO-d6, δ ppm ): 8.97 (1H, s, NH), 7.35 (1H, ddd, J =

8.4,1.5 Hz, H-5), 7.29 (1H, dd, J = 7.8, 2.0 Hz, H-3), 7.17 (1H, dd, J = 7.5, 1.6

Hz, H-6), 7.05 (1H, ddd, J = 8.1, 1.8 Hz, H-4), 6.59 (1H, dd, J = 4.5, 2.4 Hz, H-4′),

6.39 (2H, d, J = 2.1Hz, H-2′,6′), 3.77 (9H, s, OCH3); 13C NMR (DMSO-d6, δ ppm

): [178.4, 155.27] (C=N), [165.5, 150.8,135.2, 134.0, 123.5, 121.3, 116.5, 105.7,

99.7] (Ar-C), 56.5-55.7 (OCH3); GCMS (DMF, m/z, %): 343 (11, M+), 237 (100),

221 (13), 177 (21), 165 (13), 141 (8), 107 (45), 83 (6), 74 (79); Anal. Cald for

C17H17N3O3S : C, 59.46; H, 4.99; N, 12.24; S, 9.34; Found: C, 59.44; H, 4.94; N,

12.25; S, 9.34.

Page 282: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

278

2-Methoxyphenylamino-5-[1-(2,4-Dichlorophenoxy)methyl]-1,3,4-thiadiazole (90e): Yield: 61%; m.p.: 201-202°C; Rf : 0.37 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3315 (NH stretching), 3010-2893 (CH stretching), 1491 (C=N),

1615-1484 (C=C); 1H NMR (DMSO-d 6, δ ppm ): 9.05 (1H, s, NH), 7.62 (1H, s,

H-3′), 7.48 (1H, d, J = 7.5 Hz, H-5′), 7.25 (1H, d, J = 8.1 Hz, H-6′), 7.17 (1H, ddd,

J = 9.0, 1.5 Hz, H-5), 7.12 (1H, dd, J = 7.8, 1.5 Hz, H-3), 7.06 (1H, dd, J = 7.8,

1.6 Hz, H-6), 6.84 (1H, ddd, J = 7.5, 1.2 Hz, H-4), 4.69 (2H, s, CH2), 3.81 (3H, s,

OCH3); 13C NMR (DMSO-d6, δ ppm ): [164.3, 152.3] (C=N), [153.5, 149.4, 133.7,

131.8, 129.9, 125.6, 122.6, 120.4, 118.4, 118.2, 115.8] (Ar-C), 65.2 (CH2),

56.2(OCH3); GCMS (DMF, m/z, %): 282 (6, M+), 275 (100), 259 (16), 201 (24),

136 (38), 107 (12), 78 (25), 58 (4); Anal. Cald for C16H13Cl2N3O2S : C, 50.27; H,

3.43; N, 10.99; S, 8.39; Found: C, 50.24; H, 3.48; N, 10.95; S, 8.48.

2-Methoxyphenylamino-5-(3,5-Difluorophenyl)-1,3,4-thiadiazole (90f): Yield:

59%; m.p.: 178-179°C; Rf : 0.35(Petroleum ether : acetone; 6:4); IR (KBr, νmax,

cm-1): 3311 (NH stretching), 3939-2803 (CH stretching), 1508 (C=N), 1595-1384

(C=C); 1H NMR (DMSO-d6, δ ppm ): 8.98 (1H, s, NH), 7.57 (2H, dd, J = 15.5,

2.1 Hz, H-2′,6′), 7.49 (1H, m, H-4′), 7.38 (1H, ddd, J = 8.4, 1.5 Hz, H-5), 7.26

(1H, dd, J = 7.5, 1.8 Hz, H-3), 7.18 (1H, dd, J = 7.8, 1.5 Hz, H-6), 7.05 (1H, ddd,

J = 7.5, 1.2 Hz, H-4), 3.85 (3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ): [177.8,

154.6] (C=N), [164.4, 149.3, 138.0, 134.8, 123.2, 120.4, 118.5, 115.5, 112.5,

105.3] (Ar-C), 56.1 (OCH3); GCMS (DMF, m/z, %): 319 (22, M+), 213 (100), 197

(13), 221 (13), 171 (24), 165 (21), 153 (23), 107 (45), 74 (14); Anal. Cald for

C15H11F2N3OS : C, 56.42; H, 3.47; N, 13.16; S, 10.04; Found: C, 56.45; H, 3.46;

N, 13.10; S, 10.11.

4-Methoxyphenylamino-5-(3,4,5-trimethoxyphenethyl)-1,3,4-thiadiazole (90g): Yield: 55%; m.p.: 197-198°C; Rf : 0.37 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3206 (NH stretching), 3008-2870 (CH stretching), 1523 (C=N),

1594-1481 (C=C); 1H NMR (DMSO-d6, δ ppm ): 9.12 (1H, s, NH), 7.30 (1H, d, J

Page 283: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

279

= 7.8 Hz, H-3,5), 7.11 (1H, d, J = 7.5 Hz, H-2,6), 6.70 (1H, d, J = 1.8 Hz, H-2′,6′),

3.89-3.67 (12H, s, OCH3), 2.78-2.59 (4H, m, CH2); 13C NMR (DMSO-d6, δ ppm ):

[169.1, 153.9] (C=N), [151.7, 151.1, 137.2, 136.4, 133.8, 119.6, 116.3, 106.0]

(Ar-C), 57.0-56.6 (OCH3), 36.4 (CH2), 33.6 (CH2); GCMS (DMF, m/z, %): 401

(12, M+), 295 (100), 279 (13), 235 (6), 221 (13), 165 (35), 156 (18), 107 (25), 58

(7); Anal. Cald for C20H23N3O4S : C, 59.83; H, 5.77; N, 10.47; S, 7.99; Found: C,

59.87; H, 5.73; N, 10.38; S, 8.14.

4-Methoxyphenylamino-5-(3,5-dimethoxyphenyl)-1,3,4-thiadiazole (90h): Yield: 57%; m.p.: 188-189°C; Rf : 0.35 (Petroleum ether : acetone; 6:4); IR (KBr,

νmax, cm-1): 3191 (NH stretching), 3031-2913 (CH stretching), 1503 (C=N), 1600-

1484 (C=C); 1H NMR (DMSO-d6, δ ppm ): 9.05 (1H, s, NH), 7.53 (1H, d, J = 9.0

Hz, H-3,5), 7.24 (1H, d, J = 7.5Hz, H-2,6), 6.72 (1H, dd, J = 4.5, 2.4 Hz, H-4′),

6.51 (2H, d, J = 2.1 Hz, H-2′,6′), 3.71-3.61 (9H, s, OCH3); 13C NMR (DMSO-d6, δ

ppm ): [178.4, 153.4] (C=N), [164.2, 151.2, 136.8, 136.2, 118.0, 117.7, 105.5,

101.7] (Ar-C), 56.0-55.4 (OCH3); GCMS (DMF, m/z, %): 343 (25, M+), 237

(100), 221 (9), 177 (18), 165 (21), 141 (18), 107 (40), 83 (26), 74 (33); Anal. Cald

for C17H17N3O3S : C, 59.46; H, 4.99; N, 12.24; S, 9.34; Found: C, 59.49; H, 4.94;

N, 12.29; S, 9.32.

4-Methoxyphenylamino-5-[1-(2,4-dichlorophenoxy)methyl]-1,3,4-thiadiazole (90i): Yield: 55%; m.p.: 145-146°C; Rf : 0.37 (Petroleum ether : acetone; 6:4); IR

(KBr, νmax, cm-1): 3299 (NH stretching), 3038-2945 (CH stretching), 1508 (C=N),

1594-1381 (C=C); 1H NMR (DMSO-d 6, δ ppm ): 8.73 (1H, s, NH), 7.51 (1H, s,

H-3′), 7.48 (1H, d, J = 7.8 Hz, H-5′), 7.31 (1H, d, J = 8.1 Hz, H-6′), 7.17 (1H, d, J

= 9.0, 1.5 Hz, H-3,5), 7.12 (1H, d, J = 7.8, 1.5 Hz, H-2,6), 4.51 (2H, s, CH2),

3.84(3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ): [177.2, 153.3](C=N), [156.5,

152.4, 136.4, 131.8, 129.9, 125.6, 118.6, 118.3, 115.3](Ar-C), 66.2(CH2),

56.8(OCH3); GCMS (DMF, m/z, %): 282 (16, M+), 275(100), 259(10), 201(21),

Page 284: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

280

136(45), 107(21), 78(19), 58(14); Anal. Cald for C16H13Cl2N3O2S : C, 50.27; H,

3.43; N, 10.99; S, 8.39; Found: C, 50.24; H, 3.44; N, 10.97; S, 8.36.

4-Methoxyphenylamino-5-(3,5-difluorophenyl)-1,3,4-thiadiazole(90j): Yield:

54%; m.p.: 111-112°C; Rf : 0.37 (Petroleum ether : acetone; 6:4); IR (KBr, νmax,

cm-1): 3401(NH stretching), 3058-2870 (CH stretching), 1531(C=N), 1597-

1394(C=C); 1H NMR (DMSO-d6, δ ppm ): 9.04(1H, s, NH), 7.64(2H, ddd, J =

15.5,2.1 Hz, H-2′,6′), 7.51(1H, m, H-4′), 7.28(1H, d, J = 7.8 Hz, H-3,5), 7.07(1H,

d, J = 7.5 Hz, H-2,6), 3.76(3H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ): [178.6,

154.6](C=N), [166.2, 152.0, 137.7, 137.0, 121.2, 116.5, 116.5, 105.3](Ar-C),

56.6(OCH3); GCMS (DMF, m/z, %): 319 (19, M+), 213(100), 197(28), 221(34),

171(24), 165(39), 153(12), 107(33), 74(29); Anal. Cald for C15H11F2N3OS : C,

56.42; H, 3.47; N, 13.16; S, 10.04; Found: C, 56.45; H, 3.42; N, 13.14; S, 10.14.

7.10 General procedure for the synthesis of Indolinones

A mixture of haloisatin (0.01 moles) was refluxed with an equimolar

amount of the appropriate hydrazide (0.01 moles) in ethanol (10 mL) for 5 hours

and left to cool. The precipitated solid was filtered, washed with water, dried and

crystallized from the appropriate solvent.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-3,5-difluorobenzohydrazide (91a):

Yield: 89%; m.p: 178-179 oC; IR (KBr, νmax, cm-1): 3281 (NH stretching), 3084

(sp2 CH stretching), 1710 (C=O), 1677 (C=N), 1623, 1460 (C=C); 1H NMR

(DMSO-d6, δ ppm ): 11.78(1H, s, NH), 10.98 (1H, s, NH), 8.11 (1H, s, H-4),

7.68-7.43 (4H, m, H-6,2′,4′,6′), 6.93 (1H, d, J = 8.7Hz, H-7); 13C NMR (DMSO-d

6, δ ppm ): 171.3, 164.7, 164.2, 143.3, 141.7, 136.7, 132.8, 127.4, 127.1, 126.0,

116.8, 112.9, 108.0; EIMS (m/z, %): 335 (48, M+), 307 (14), 290 (2), 287 (1), 250

(2), 244 (2), 216 (1), 194 (100), 166 (53), 141 (91), 113 (56), 102 (10), 75 (7), 63

(6), 51 (2); Anal. Cald for C15H8ClF2N3O2: C, 53.67; H, 2.40; N, 12.52; Found: C,

53.62; H, 2.45; N, 12.58.

Page 285: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

281

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2,5-difluorobenzohydrazide (91b):

Yield: 88%; m.p: 165-166 oC; IR (KBr, νmax, cm-1): 3412-3237 (NH stretching),

3115 (sp2 CH stretching), 1719 (C=O), 1622 (C=N), 1308 (C-O); 1H NMR

(DMSO-d6, δ ppm ): 11.44(1H, s, NH), 10.96(1H, s, NH), 8.13(1H, s, H-4), 7.72-

7.41(3H, m, H-3′,4′,6′), 7.08-6.79(2H, m, H-6,7); 13C NMR (DMSO-d6, δ ppm ):

170.5, 162.2, 160.7, 159.1, 145.3, 133.7, 132.8, 131.4, 129.1, 127.5, 124.0,

121.9, 120.8, 118.5, 116.9; EIMS (m/z, %): 335 (24, M+), 307 (24), 291 (1), 290

(1), 250 (1), 232 (2), 224 (1), 194 (55), 166 (37), 141 (100), 113 (44), 102 (12),

75 (7), 63 (7), 51 (1); Anal. Cald for C15H8ClF2N3O2: C, 53.67; H, 2.40; N, 12.52;

Found: C, 53.66; H, 2.43; N, 12.53.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2,6-difluorobenzohydrazide (91c):

Yield: 84%; m.p: 212-213oC; IR (KBr, νmax, cm-1): 3419-3232 (NH stretching),

3118 (sp2 CH stretching), 1713 (C=O), 1627 (C=N), 1278 (C-O); 1H NMR

(DMSO-d 6, δ ppm ): 11.45 (1H, s, NH), 10.71 (1H, s, NH), 8.00 (1H, s, H-4),

7.54-7.31 (3H, m, H-3′,4′,6′), 6.76 (2H, d, J = 14.4 Hz, H-6,7); 13C NMR (DMSO-

d6, δ ppm) : 169.8, 163.7, 160.2, 144.0, 136.9, 133.4, 131.9, 131.2, 130.1, 124.5,

120.2, 115.5, 110.5; EIMS (m/z, %): 335 (18, M+), 307 (9), 281 (7), 207 (13), 194

(72), 166 (48), 141 (100), 113 (94), 102 (17), 75 (22), 63 (37), 44 (24), 32 (17);

Anal. Cald for C15H8ClF2N3O2: C, 53.67; H, 2.40; N, 12.52; Found: C, 53.68; H,

2.48; N, 12.55.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-3,4-dimethoxybenzohydrazide

(91d): Yield: 83%; m.p: 145-146oC; IR (KBr, νmax, cm-1): 3252 (NH stretching),

3101 (sp2 CH stretching), 1711 (C=O), 1652 (C=N), 1594-1463 (C=C), 1350 (C-

O); 1H NMR (DMSO-d6, δ ppm ): 11.49 (1H, s, NH), 10.24 (1H, s, NH), 7.60 (1H,

d, J = 2.0 Hz, H-4), 7.50-7.42 (3H, m, H-6,2′,6′), 7.19 (1H, d, J = 8.4 Hz, H-7),

6.99 (1H, d, J = 8.4 Hz, H-5′), 3.87-3.85 (6H, s, OCH3); 13C NMR (DMSO-d 6, δ

ppm ): 169.3, 163.4, 154.2, 149.9, 145.2, 133.5, 131.3, 130.8, 129.1, 128.0,

Page 286: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

282

124.0, 121.3, 120.5, 116.5, 113.2, 56.42 (OCH3); EIMS (m/z, %):359 (3, M+), 330

(3), 279 (21), 261 (1), 194 (2), 182 (3), 167 (40), 149 (100), 113 (9), 104 (7), 71

(9), 57 (10), 55 (4); Anal. Cald for C17H14ClN3O4: C, 56.75; H, 3.92; N, 11.68;

Found: C, 56.70; H, 3.98; N, 11.63.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-3,5-dimethoxybenzohydrazide

(91e): Yield: 85%; m.p: 112-113oC; IR (KBr, νmax, cm-1): 3249 (NH stretching),

3185 (sp2 CH stretching), 1670 (C=O), 1681 (C=N), 1598-1461 (C=C), 1307 (C-

O); 1H NMR (DMSO-d6, δ ppm ): 12.42 (1H, s, NH), 11.33 (1H, s, NH), 7.52 (1H,

d, J = 2.4 Hz, H-4), 7.39 (2H, dd, J = 2.0 Hz, H-2′,6′), 7.32 (1H, dd, J = 6.8, 2.4

Hz, H-6), 7.16 (1H, d, J = 8.8Hz, H-7), 6.92 (1H, dd, J = 2.0 Hz, H-4′), 3.35 (6H,

s, OCH3); 13C NMR (DMSO-d 6, δ ppm ): 170.4, 165.2, 162.2, 145.2, 137.0,

135.5, 133.3, 131.8, 124.1, 120.5, 119.5, 105.2, 56.7 (OCH3); EIMS (m/z, %):

359 (15, M+), 331 (14), 288 (24), 264 (2), 194 (5), 193 (3), 165 (100), 137 (25),

122 (35), 107 (9), 102 (8), 77 (7), 63 (5), 51 (2); Anal. Cald for C17H14ClN3O4: C,

56.75; H, 3.92; N, 11.68; Found: C, 56.76; H, 3.94; N, 11.67.

(E)--N′-(5-Chloro-2-oxoindolin-3-ylidene)-2,4-dimethoxybenzohydrazide

(91f): Yield: 80%; m.p: 201-202oC; IR (KBr, νmax, cm-1): 3345 (NH stretching),

3138 (sp2 CH stretching), 1715 (C=O), 1652 (C=N), 1594-1461 (C=C), 1350 (C-

O); 1H NMR (DMSO-d6, δ ppm ): 10.96 (1H, s, NH), 8.30 (1H, s, NH), 7.61 (1H,

d, J = 5.2 Hz, H-4), 7.44 (2H, dd, J = 14.2, 2.4 Hz, H-6,5′), 7.35 (2H, d, J = 13.2

Hz, H-7,6′), 6.94 (1H, d, J = 2.4 Hz, H-3′), 3.36 (6H, s, OCH3); 13C NMR (DMSO-

d6, δ ppm ): 169.0, 164.2, 161.6, 145.4, 134.5, 133.3, 132.0, 129.1, 124.1, 120.3,

118.5, 111.2, 107.2, 101.3, 56.4 (OCH3); EIMS (m/z, %): 359 (5, M+), 331 (8),

309 (2), 281 (3), 264 (3), 194 (4), 165 (100), 138 (8), 122 (15), 111 (9), 71 (18),

57 (21), 43 (10); Anal. Cald for C17H14ClN3O4: C, 56.75; H, 3.92; N, 11.68;

Found: C, 56.64; H, 3.95; N, 11.68.

Page 287: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

283

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2,6-dimethoxybenzohydrazide

(91g): Yield: 85%; m.p: 155-156oC; IR (KBr, νmax, cm-1): 3255 (NH stretching),

3132 (sp2 CH stretching) 1712 (C=O),1615 (C=N), 1592-1463 (C=C), 1342 (C-

O); 1H NMR (DMSO-d6, δ ppm ): 11.48 (1H, s, NH), 7.95 (1H, s, NH), 7.59 (1H,

d, J = 3.6 Hz, H-4), 7.44 (1H, dd, J = 16.8, 4.0 Hz, H-6), 7.09-6.81 (1H, m, H-

7,3′,4′,5′), 3.43 (6H, s, OCH3); 13C NMR (DMSO-d 6, δ ppm ): 169.4, 163.2,

160.2, 145.6, 135.2, 133.3, 132.8, 124.1, 121.0, 119.5, 108.5, 106.2, 56.0

(OCH3); EIMS (m/z, %): 359 (18, M+), 331 (17), 300 (2), 288 (1), 194 (5), 165

(100), 137 (24), 122 (29), 107 (6), 77 (4), 63 (4), 51 (2); Anal. Cald for

C17H14ClN3O4: C, 56.75; H, 3.92; N, 11.68; Found: C, 56.74; H, 3.95; N, 11.73.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2-(4-chlorophenyl)acetohydrazide

(91h): Yield: 89%; m.p: 139-140oC; IR (KBr, νmax, cm-1): 3216 (NH stretching),

3153 (sp2 CH stretching), 1727 (C=O),1685 (C=N), 1596-1470 (C=C), 1311 (C-

O); 1H NMR (DMSO-d6, δ ppm ): 10.93 (1H, s, NH), 8.29 (1H, s, NH), 7.94 (1H,

s, H-4), 7.56 (1H, d, J = 8.1 Hz, H-7), 7.21 (1H, d, J = 9.9 Hz, H-6), 6.89 (2H, d, J

= 12.3 Hz, H-2′,6′), 6.62 (2H, d, J = 13.5 Hz, H-3′,5′), 4.04 (2H, s, CH2); 13C NMR

(DMSO-d6, δ ppm ): 171.1, 166.4, 145.3, 134.5, 131.3, 130.8, 130.2, 129.1,

124.0, 120.5, 46.4 (CH2). EIMS (m/z, %): 347 (16, M+), 319 (5), 304 (2), 279 (3),

211 (3), 194 (43), 181 (205), 166 (26), 153 (56), 125 (100), 111 (13), 89 (20), 73

(22), 63 (11), 57 (10); Anal. Cald for C16H11Cl2N3O2: C, 55.19; H, 3.18; N, 12.07;

Found: C, 55.25; H, 3.18; N, 12.13.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2-(4-Flourophenyl)acetohydrazide

(91i) : Yield: 79%; m.p: 188-189 oC; IR (KBr, νmax, cm-1): 3262 (NH stretching),

3146 (sp2 CH stretching), 1720 (C=O), 1687 (C=N), 1602-1507 (C=C), 1313

(C=O); 1H NMR (DMSO-d6, δ ppm ): 11.35 (1H, s, NH), 10.93 (1H, s, NH), 7.66

(1H, s, H-4), 7.37 (2H, d, J = 8.0 Hz, H-6,7), 7.12 (2H, d, J = 8.4 Hz, H-2′,6′),

6.92 (2H, d, J = 8.4Hz, H-3′,5′), 4.02 (2H, s, CH2); 13C NMR (DMSO-d6, δ ppm ):

172.6, 165.3, 162.2, 146.4, 133.2, 132.0, 131.8, 129.9, 124.0, 120.1, 116.5,

Page 288: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

284

115.2, 43.4 (CH2); EIMS (m/z, %): 331 (15, M+), 303 (6), 279 (2), 222 (1), 194

(46), 181 (2), 166 (24), 153 (9), 138 (21), 125 (12), 109 (100), 102 (11), 83 (12),

75 (7), 63 (4), 44 (1); Anal. Cald for C16H11ClFN3O2: C, 57.93; H, 3.34; N, 12.67;

Found: C, 57.98; H, 3.37; N, 12.62.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-3-(3,4,5-trimethoxyphenyl)-

propionatehydrazide (91j): Yield: 86%; m.p: 150-151 oC; IR (KBr, νmax, cm-1):

3223 (NH stretching), 3107 (sp2 CH stretching), 1739 (C=O), 1614 (C=N), 1463

(C=C), 1342 (C-O); 1H NMR (DMSO-d6, δ ppm ): 10.42 (1H, s, NH), 8.45 (1H, s,

NH), 7.55 (1H, s, H-4), 7.39 (1H, dd, J = 8.4,2.0 Hz, H-6), 6.93 (1H, d, J = 8.0

Hz, H-7), 6.58 (1H, s, H-2′,6′), 3.75-3.35 (12H, s, OCH3), 2.88 (2H, s, CH2), 2.72

(2H, s, CH2); 13C NMR (DMSO-d6, δ ppm ): 169.1, 168.4, 152.5, 145.8, 137.2,

134.5, 133.3, 132.0, 130.5, 129.8, 124.2, 120.5, 106.5, 40.2 (CH2), 36.4 (CH2);

EIMS (m/z, %): 417 (32, M+), 400 (4), 372 (8), 358 (3), 341 (2), 313 (2), 236 (12),

223 (7), 194 (18), 181 (100), 166 (16), 138 (12), 102 (7), 77 (6), 65 (3), 51 (2);

Anal. Cald for C20H20ClN3O5: C, 57.49; H, 4.82; N, 10.06; Found: C, 57.46; H,

4.89; N, 10.00.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-3-(4-methoxyphenyl)propionate-

hydrazide (91k): Yield: 83%; m.p: 198-199 oC; IR (KBr, νmax, cm-1): 3256 (NH

stretching), 3074 (sp2 CH stretching), 1746 (C=O), 1680 (C=N), 1645-1486

(C=C), 1391 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 10.89 (1H, s, NH), 8.27 (1H, s,

NH), 7.94 (1H, s, H-4), 7.40 (1H, dd, J = 8.4, 1.6 Hz, H-6), 7.18 (1H, dd, J = 6.8,

2.0 Hz, H-2′,6′), 6.88 (1H, d, J = 8.4 Hz, H-7), 6.84 (1H, dd, J = 6.4, 1.6 Hz, H-

3′,5′), 3.35 (3H, s, OCH3), 2.88 (2H, s, CH2), 2.72 (2H, s, CH2); 13C NMR (DMSO-

d6, δ ppm ): 169.4, 168.2, 158.5, 144.8, 133.5, 133.4, 132.4, 130.5, 129.8, 128.5,

124.2, 120.5, 115.3, 39.2 (CH2), 36.0 (CH2); EIMS (m/z, %): 357 (18, M+), 340

(7), 312 (17), 298 (4), 277 (5), 194 (8), 176 (12), 166 (11), 121 (100), 91 (13), 77

(9), 65 (3), 51 (1); Anal. Cald for C18H16ClN3O3: C, 60.42; H, 4.51; N, 11.74;

Found: C, 60.46; H, 4.41; N, 11.75.

Page 289: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

285

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-3-(4-bromophenoxy)acetato-

hydrazide (91l): Yield: 85%; m.p: 205-206oC; IR (KBr, νmax, cm-1): 3286(NH

stretching), 3074 (sp2 CH stretching), 1746 (C=O), 1645 (C=N), 1486 (C=C),

1391 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.61 (1H, s, NH), 10.95 (1H, s, NH),

7.94 (1H, s, H-4), 7.46 (3H, d, J = 9.6 Hz, H-7, 3′,5′), 6.92 (3H, dd, J = 8.7 Hz, H-

6,2′,6′), 5.16 (2H, s, CH2); 13C NMR (DMSO-d6, δ ppm ): 164.6, 162.7, 157.6,

143.0, 132.5, 126.3, 126.2, 117.2, 116.6, 65.9 (CH2); EIMS (m/z, %): 409 (48,

M+,

Br81), 407 (38, M+,

Br79), 340 (7), 312 (17), 298 (4), 277 (5), 194 (8), 176 (12),

166 (11), 121 (100), 91 (13), 77 (9), 65 (3), 51 (1); Anal. Cald for

C16H11BrClN3O3: C, 47.03; H, 2.71; N, 10.28; Found: C, 47.07; H, 2.79; N, 10.23.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2-(2,4-dichlorophenoxy)aceto-

hydrazide(91m): Yield: 79%; m.p: 191-192oC; IR (KBr, νmax, cm-1): 3184 (NH

stretching), 3052 (sp2 CH stretching), 1726 (C=O), 1625 (C=N), 1489 (C=C),

1371 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.48 (1H, s, NH), 7.94 (1H, s, NH),

7.58 (1H, s, H-4), 7.43 (2H, d, J = 8.4, 2.4 Hz, H-6,5′), 6.98 (1H, d, J = 8.0 Hz, H-

3′), 6.81 (2H, d, J = 8.0 Hz, H-7,6′), 3.82 (2H, s, CH2); 13C NMR (DMSO-d 6, δ

ppm ): 172.6, 168.7, 153.6, 145.0, 133.2, 132.3, 131.2, 129.2, 124.6, 120.2,

118.5, 67.9 (CH2); EIMS (m/z, %): 397 (02, M+), 362 (1), 264 (100), 231 (6), 219

(2), 194 (14), 166 (16), 162 (10), 138 (13), 111 (7), 102 (7), 69 (13), 41 (3). Anal.

Cald for C16H10Cl3N3O3: C, 48.21; H, 2.53; N, 10.54; Found: C, 48.25; H, 2.59; N,

10.50.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2-(2,4-dichlorophenoxy)propionato-

hydrazide (91n): Yield: 82%; m.p: 116-117 oC; IR (KBr, νmax, cm-1): 3226 (NH

stretching), 3096 (sp2 CH stretching), 1735 (C=O), 1615 (C=N), 1473 (C=C),

1206 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.34 (1H, s, NH), 10.90 (1H, s, NH),

7.95 (1H, s, H-4), 7.79 (1H, s, H-4,3′), 7.60-7.36 (2H, m, H-6,5′), 6.78 (1H, d, J =

12.3 Hz, H-7,6′), 5.38 (1H, s, CH), 1.59 (2H, s, CH3); 13C NMR (DMSO-d6, δ ppm

Page 290: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

286

): 173.6, 169.7, 151.6, 144.7, 133.4, 132.3, 131.2, 129.2, 125.6, 123.5, 119.9,

117.8, 75.9 (CH) 15.5 (CH3); EIMS (m/z, %): 409 (4, M+), 394 (5), 366 (8), 352

(8), 323 (9), 295 (12), 253 (16), 239 (17), 197 (20), 183 (21), 155 (23), 113 (33),

99 (38), 85 (100), 71 (97), 57 (78), 43 (18); Anal. Cald for C17H12Cl3N3O3: C,

49.48; H, 2.93; N, 10.18; Found: C, 49.46; H, 2.99; N, 10.15.

(E)-N′-(5-Chloro-2-oxoindolin-3-ylidene)-2-(2,4-dichlorophenoxy)butyrato-

hydrazide (91o): Yield: 89%; m.p: 133-134 oC; IR (KBr, νmax, cm-1): 3451 (NH

stretching), 3091 (sp2 CH stretching), 1740 (C=O), 1680 (C=N), 1448, 1429

(C=C), 1384 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 10.97 (1H, s, NH), 8.31 (1H, s,

NH), 7.63-6.90 (6H, m, Ar-H), 5.29 (1H, s, CH2), 3.57 (1H, s, CH2), 2.51 (1H, s,

CH2); 13C NMR (DMSO-d 6, δ ppm ): 168.6, 166.7, 151.6, 146.1, 133.5, 132.3,

131.2, 130.2, 128.6, 124.2, 120.6, 117.8, 69.9 (CH2), 35.5 (CH2), 25.9 (CH2); EIMS (m/z, %): 427 (2, M+), 397 (1), 362 (1), 264 (100), 231 (4), 194 (14), 166

(15), 162 (9), 138 (3), 111 (7), 102 (7), 69 (13), 41 (4); Anal. Cald for

C18H14Cl3N3O3: C, 50.67; H, 3.31; N, 9.85; Found: C, 50.61; H, 3.32; N, 9.80.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-3,5-difluorobenzohydrazide (92a):

Yield: 91%; m.p: 158-159 oC; IR (KBr, νmax, cm-1): 3228 (NH stretching), 3042

(sp2 CH stretching), 2996-2834, 1708 (C=O), 1595 (C=N), 1351 (C-O); 1H NMR

(DMSO-d 6, δ ppm ): 11.53 (1H, s, NH), 10.49 (1H, s, NH), 7.76 (1H, s, H-4),

7.62-7.45 (4H, m, H-6,2′,4′,6′), 6.97 (1H, d, J = 8.7 Hz, H-7); 13C NMR (DMSO-

d6, δ ppm ): 169.3, 165.7, 163.2, 145.3, 138.7, 134.7, 132.8, 125.4, 121.8, 119.8,

104.9; EIMS (m/z, %): 381 (25, M+, Br81), 379 (25, M+

, Br79), 353 (10, Br81), 351

(10, Br79), 301 (1), 240 (61, Br81), 238 (61, Br79), 212 (34, Br81), 210 (35, Br79),

184 (12, Br81), 182 (12, Br79), 141 (100), 113 (69), 103 (22), 63 (10); Anal. Cald

for C15H8BrF2N3O2: C, 47.39; H, 2.12; N, 11.05; Found: C, 47.38; H, 2.15; N,

11.02.

Page 291: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

287

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2,5-difluorobenzohydrazide (92b):

Yield: 88%; m.p: 139-140oC; IR (KBr, νmax, cm-1): 3231 (NH stretching), 3052 (sp2

CH stretching), 2983-2856, 1705 (C=O), 1625 (C=N), 1590-1359 (C=C), 1316

(C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.23 (1H, s, NH), 10.79 (1H, s, NH), 7.81

(1H, s, H-4), 7.72-7.54 (3H, m, H-3′,4′,6′), 7.16-6.72 (2H, m, H-6,7); 13C NMR

(DMSO-d 6, δ ppm ): 169.2, 164.2, 159.2, 158.1, 144.3, 132.7, 130.8, 130.4,

127.5, 124.0, 121.9, 120.8, 119.3, 118.5, 116.9; EIMS (m/z, %): 381 (21, M+, Br81), 379 (21, M+, Br79), 353 (17, Br81), 351 (17, Br79), 301 (4), 273 (5), 240 (43,

Br81), 238 (43, Br79), 212 (22 Br81), 210 (22, Br79), 184 (5, Br81), 182 (5, Br79), 141

(100), 113 (39), 103 (12), 63 (6); Anal. Cald for C15H8BrF2N3O2: C, 47.39; H,

2.12; N, 11.05; Found: C, 47.32; H, 2.11; N, 11.03.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2,6-difluorobenzohydrazide (92c):

Yield:85%; m.p: 184-185oC; IR (KBr, νmax, cm-1): 3227 (NH stretching), 3025 (sp2

CH stretching), 2987-2850, 1715 (C=O), 1651 (C=N), 1597-1349 (C=C), 1326

(C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.21 (1H, s, NH), 10.41 (1H, s, NH), 7.74

(1H, s, H-4), 7.54-7.31 (3H, m, H-3′,4′,6′), 6.71 (2H, d, J = 14.4 Hz, H-6,7); 13C

NMR (DMSO-d 6, δ ppm ): 168.3, 164.2, 161.2, 145.2, 136.2, 135.7, 133.2,

132.2, 120.2, 115.5, 111.5; EIMS (m/z, %): 381 (4, M+, Br81), 379 (4, M+, Br79),

353 (54, Br81), 351 (54, Br79), 301 (34), 273 (4), 240 (23, Br81), 238 (23, Br79),

212 (12, Br81), 210 (12, Br79), 184 (4, Br81), 182 (4, Br79), 141 (100), 113 (46), 103

(2), 63 (2); Anal. Cald for C15H8BrF2N3O2: C, 47.39; H, 2.12; N, 11.05; Found: C,

47.37; H, 2.19; N, 11.09.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-3,4-dimethoxybenzohydrazide

(92d): Yield: 87%; m.p: 121-122 oC; IR (KBr, νmax, cm-1): 3223 (NH stretching),

3054 (sp2 CH stretching), 2992-2836, 1742 (C=O), 1678 (C=N), 1536 (C=C),

1308 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.33 (1H, s, NH), 10.49 (1H, s, NH),

7.57 (1H, d, J = 2.4 Hz, H-4), 7.45-7.39 (3H, m, H-6,2′,6′), 7.11 (1H, d, J = 8.4

Hz, H-7), 6.93 (1H, d, J = 8.4 Hz, H-5′), 3.89-3.82 (6H, s, OCH3); 13C NMR

Page 292: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

288

(DMSO-d6, δ ppm ): 168.3, 164.4, 153.2, 151.2, 145.2, 134.1, 133.5, 131.3,

127.9, 123.4, 120.3, 119.5, 117.2, 113.4, 56.5 (OCH3); EIMS (m/z, %): 405 (22,

M+, Br81), 403 (22, M+, Br79), 379 (8, Br81), 377 (8, Br79), 351 (5), 284 (2), 240 (19,

Br81), 238 (19, Br79), 178 (3), 165 (100), 137 (7), 122 (19), 77 (15); Anal. Cald for

C17H14BrN3O4: C, 50.51; H, 3.49; N, 10.40; Found: C, 50.55; H, 3.48; N, 10.43.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-3,5-dimethoxybenzohydrazide

(92e): Yield: 80%; m.p: 204-205oC; IR (KBr, νmax, cm-1): 3228 (NH stretching),

3065 (sp2 CH stretching), 2999-2835, 1747 (C=O), 1678 (C=N), 1535 (C=C),

1306 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.56 (1H, s, NH), 10.02 (1H, s, NH),

7.70 (1H, s, H-4), 7.56 (1H, dd, J = 8.4, 2.0 Hz, H-6), 6.98 (2H, d, J = 2.4 Hz, H-

2′,6′), 6.93 (1H, d, J = 8.0 Hz, H-7), 6.82 (1H, dd, J = 2.0 Hz, H-4′), 3.83 (6H, s,

OCH3); 13C NMR (DMSO-d 6, δ ppm ): 168.6, 162.6, 160.7, 141.4, 138.8, 135.2,

133.9, 123.1, 121.2, 119.3, 105.1, 104.3, 55.6 (OCH3). EIMS (m/z, %): 405 (15,

M+, Br81), 403 (15, M+, Br79), 379 (11, Br81), 377 (11, Br79), 351 (2), 284 (1), 240

(9, Br81), 238 (9, Br79), 178 (23), 165 (100), 137 (17), 122 (16), 77 (5). Anal. Cald

for C17H14BrN3O4: C, 50.51; H, 3.49; N, 10.40; Found: C, 50.55; H, 3.43; N,

10.41.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2,4-dimethoxybenzohydrazide

(92f): Yield: 86%; m.p: 167-168 oC; IR (KBr, νmax, cm-1): 3486-3211 (NH

stretching), 3075 (sp2 CH stretching), 2839, 1717 (C=O), 1652 (C=N), 1492

(C=C), 1238 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.54 (1H, s, NH), 10.24 (1H,

s, NH), 7.72 (1H, s, H-4), 7.53 (1H, dd, J = 8.4, 2.0 Hz, H-6), 7.24 (1H, d, J = 8.4

Hz, H-6′), 6.99 (1H, d, J = 8.0 Hz, H-7), 6.70 (1H, dd, J = 7.8, 2.0 Hz, H-5′), 6.67

(1H, d, J = 2.1 Hz, H-3′), 3.88 (6H, s, OCH3); 13C NMR (DMSO-d6, δ ppm ):

168.2, 163.5, 161.7, 141.4, 134.2, 133.3, 128.1, 124.2, 121.2, 111.8, 106.3,

101.1, 55.3 (OCH3); EIMS (m/z, %): 405 (5, M+, Br81), 403 (5, M+, Br79), 377 (6,

Br81), 375 (6, Br79), 325 (2), 297 (3), 240 (1), 165 (100), 122 (7), 107 (6), 77 (3);

Anal. Cald for C17H14BrN3O4: C, 50.51; H, 3.49; N, 10.40; Found: C, 50.55; H,

3.45; N, 10.49.

Page 293: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

289

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2,6-dimethoxybenzohydrazide

(92g): Yield: 88%; m.p: 128-129 oC; IR (KBr, νmax, cm-1): 3416-3311 (NH

stretching), 3072 (sp2 CH stretching), 2839, 1717 (C=O), 1606 (C=N), 1592

(C=C), 1308 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.78 (1H, s, NH), 10.49 (1H,

s, NH), 7.85 (1H, s, H-4), 7.62 (1H, dd, J = 8.0, 2.1 Hz, H-6), 7.41 (1H, d, J = 8.4

Hz, H-6′), 7.05 (1H, d, J = 8.0 Hz, H-7), 6.79 (1H, dd, J = 7.8, 2.0 Hz, H-5′), 6.71

(1H, d, J = 2.1 Hz, H-3′), 3.80 (6H, s, OCH3); 13C NMR (DMSO-d 6, δ ppm ):

169.6, 164.5, 161.7, 145.4, 134.2, 133.3, 124.7, 121.2, 119.8, 106.3, 56.4

(OCH3); EIMS (m/z, %): 405 (5, M+, Br81), 403 (6, M+, Br79), 377 (4, Br81), 375 (4,

Br79), 325 (1), 297 (2), 240 (1), 165 (100), 122 (9), 107 (4), 77 (2), 57 (2); Anal.

Cald for C17H14BrN3O4: C, 50.51; H, 3.49; N, 10.40. Found: C, 50.50; H, 3.44; N,

10.47.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2-(4-chlorophenyl)acetohydrazide

(92h): Yield: 85%; m.p: 137-138oC; IR (KBr, νmax, cm-1): 3577-3110 (NH

stretching), 3011 (sp2 CH stretching), 1636 (C=O), 1611 (C=N), 1561 (C=C),

1254 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 11.81 (1H, s, NH), 10.29 (1H, s, NH),

7.99 (1H, s, H-4), 7.69 (1H, d, J = 8.1 Hz, H-7), 7.45 (1H, d, J = 9.9 Hz, H-6),

7.09 (2H, d, J = 12.3 Hz, H-2′,6′), 7.01 (2H, d, J = 13.5 Hz, H-3′,5′), 4.14 (2H, s,

CH2); 13C NMR (DMSO-d 6, δ ppm ): 171.4, 164.3, 145.2, 134.0, 133.9, 133.5,

131.3, 130.8, 124.1, 121.0, 119.0, 56.4 (OCH3), 49.2 (CH2); EIMS (m/z, %): 347

(16, M+), 319 (5), 304 (2), 279 (3), 211 (3), 194 (43), 181 (20), 166 (26), 153 (56),

125 (100), 111 (13), 89 (20), 73 (22), 63 (11), 57 (10); Anal. Cald for

C16H11BrClN3O2: C, 48.94; H, 2.82; N, 10.70; Found: C, 48.92; H, 2.84; N, 10.75.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2-(4-Flourophenyl)aceto-

hydrazide(92i): Yield: 81%; m.p: 157-158 oC; IR (KBr, νmax, cm-1): 3547-3410

(NH stretching), 1636 (C=O), 1600 (C=N), 1561 (C=C), 1231 (C-O); 1H NMR

(DMSO-d 6, δ ppm ): 11.19 (1H, s, NH), 10.81 (1H, s, NH), 7.79 (1H, s, H-4),

Page 294: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

290

7.54 (2H, d, J = 8.0 Hz, H-6,7), 7.09 (2H, d, J = 8.4 Hz, H-2′,6′), 6.98 (2H, d, J =

8.4 Hz, H-3′,5′), 4.14 (2H, s, CH2); 13C NMR (DMSO-d 6, δ ppm ): 168.7, 165.5,

160.2, 145.5, 134.5, 133.8, 133.2, 130.1, 130.0, 124.1, 120.3, 119.5, 115.2, 56.7

(OCH3), 48.6 (CH2); EIMS (m/z, %): 375 (11, M+), 347 (8), 323(3), 266(2), 238

(49), 225(11), 210(24), 197(11), 182 (21), 169 (12), 153(100), 146(11), 127 (12),

119 (10), 107(5), 88 (1). Anal. Cald for C16H11BrFN3O2: C, 51.08; H, 2.95; N,

11.17; Found: C, 51.12; H, 2.94; N, 11.12.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-3-(3,4,5-trimethoxyphenyl)-

propionatehydrazide (92j): Yield: 84%; m.p: 101-102 oC; IR (KBr, νmax, cm-1):

3272 (NH stretching), 2940, 2833, 1733 (C=O), 1654 (C=N), 1594-1460 (C=C),

1241 (C-O); 1H NMR (DMSO-d 6, δ ppm ): 10.90 (1H, s, NH), 8.37 (1H, s, NH),

7.65 (1H, s, H-4), 7.51 (1H, d, J = 7.6 Hz, H-7), 6.88 (1H, d, J = 10.4 Hz, H-6),

6.56 (1H, s, H-2′,6′), 3.74-3.36 (12H, s, OCH3), 2.87 (2H, s, CH2), 2.50 (2H, s,

CH2); 13C NMR (DMSO-d 6, δ ppm ): 169.3, 168.4, 152.6, 142.3, 137.5, 134.6,

134.0, 133.8, 123.7, 122.9, 119.5, 105.5, 55.7 (OCH3), 36.4 (CH2), 30.4 (CH2);

EIMS (m/z, %): 463 (33, M+, Br81), 461 (33., M+, Br79), 418 (7, Br81), 416 (7, Br79),

383 (5), 337 (2), 284 (1), 236 (13), 181 (100, Br81), 179 (11, Br79), 151 (4, Br81),

149 (4., Br79), 103 (4), 77 (3), 65 (2.); Anal. Cald for C20H20BrN3O5: C, 51.96; H,

4.36; N, 9.09; Found: C, 51.99; H, 4.39; N, 9.12.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-3-(4-methoxyphenyl)propionate-

hydrazide (92k): Yield: 80%; m.p: 130-131 oC; IR (KBr, νmax, cm-1): 3201 (NH

stretching), 3092, 2836, 1734 (C=O), 1648 (C=N), 1597-1511 (C=C), 1375 (C-O);

1H NMR (DMSO-d 6, δ ppm ): 10.81 (1H, s, NH), 8.42 (1H, s, NH), 7.65 (1H, s,

H-4), 7.49 (1H, d, J = 8.4 Hz, H-6,7), 7.23 (1H, d, J = 6.8 Hz, H-2′,6′), 6.91 (1H,

d, J = 6.4 Hz, H-3′,5′), 3.38 (3H, s, OCH3), 2.92 (2H, s, CH2), 2.45 (2H, s, CH2); 13C NMR (DMSO-d 6, δ ppm ): 173.8, 167.0, 152.1, 143.9, 142.3, 138.8, 137.6,

126.3, 123.2, 122.9, 121.8, 116.5, 56.5 (OCH3), 35.7 (CH2), 30.7 (CH2); EIMS

(m/z, %): 403 (15, M+, Br81), 401 (15, M+, Br79), 358 (12, Br81), 356 (12, Br79),

Page 295: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

291

277 (5), 241 (6, Br81), 239 (6, Br79), 214 (5, Br81), 212 (5, Br79), 176 (20), 121

(100), 91 (8) 77 (8), 57 (3); Anal. Cald for C18H16BrN3O3: C, 53.75; H, 4.01; N,

10.45; Found: C, 53.79; H, 4.11; N, 10.42.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-3-(4-bromophenoxy)aceto-

hydrazide (92l): Yield: 81%; m.p: 155-156 oC; IR (KBr, νmax, cm-1): 3281 (NH

stretching), 3120, 2925, 1745-1713 (C=O), 1645 (C=N), 1585 (C=C), 1287 (C-O);

1H NMR (DMSO-d 6, δ ppm ): 10.96 (1H, s, NH), 8.33 (1H, s, NH), 7.94 (1H, s,

H-4), 7.55 (3H, d, J = 8.4 Hz, H-7), 7.46 (3H, dd, J = 8.8 Hz, H-6), 7.03-6.83 (4H,

m, H-2′,3′,5′,6′), 5.15 (2H, s, CH2); 13C NMR (DMSO-d 6, δ ppm ): 163.9, 162.1,

157.0, 142.7, 134.7, 132.0, 128.2, 116.7, 114.2, 64.7 (CH2); EIMS (m/z, %): 453

(15, M+, Br81), 451 (10, M+, Br79), 375 (3, Br81), 373 (3, Br79), 356 (2), 282 (3,

Br81), 280 (4, Br79), 240 (15, Br81), 242 (15, Br79), 212 (8, Br81), 210 (8, Br79), 157

(8, Br81), 155 (8, Br79), 73 (100), 44 (24); Anal. Cald for C16H11Br2N3O3: C,

42.41; H, 2.45; N, 9.27; Found: C, 42.47; H, 2.40; N, 9.22.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2-(2,4-dichlorophenoxy)aceto-

hydrazide (92m): Yield: 88%; m.p: 171-172 oC; IR (KBr, νmax, cm-1): 3189 (NH

stretching), 2948, 1749 (C=O), 1654 (C=N), 1596 (C=C), 1298 (C-O); 1H NMR

(DMSO-d 6, δ ppm ): 10.88 (1H, s, NH), 8.37 (1H, s, NH), 7.59 (1H, d, J = 1.8 Hz,

H-4), 7.52 (1H, dd, J = 5.8, 2.2 Hz, H-6), 7.34 (1H, dd, J = 6.2, 1.8 Hz, H-5′),

7.19 (1H, d, J = 2.2 Hz, H-3′), 6.88 (1H, d, J = 8.4 Hz, H-7), 6.83 (1H, d, J = 8.4

Hz, H-6′), 3.34 (2H, s, CH2); 13C NMR (DMSO-d 6, δ ppm ): 167.6, 163.7, 152.8,

142.5, 135.9, 134.5, 129.1, 127.9, 124.2, 122.2, 116.7, 114.8, 68.2 (CH2); EIMS

(m/z, %): 445 (15, M+, Br81), 443 (33, M+, Br79), 408 (53, Br81), 406 (39, Br79),

328 (37), 282 (9, Br81), 280 (12, Br79), 240 (95, Br81), 238 (100, Br79), 212 (53,

Br81), 210 (58, Br79), 162 (42, Br81), 160 (88, Br79), 145 (73), 73 (77). Anal. Cald

for C16H10BrCl2N3O3: C, 43.37; H, 2.27; N, 9.48; Found: C, 43.47; H, 2.20; N,

9.42.

Page 296: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

292

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2-(2,4-dichlorophenoxy)-

propionatohydrazide (92n): Yield: 83%; m.p: 196-198 oC; IR (KBr, νmax, cm-1):

3557-3419 (NH stretching), 1636 (C=O), 1601 (C=N), 1561 (C=C), 1238 (C-O);

1H NMR (DMSO-d 6, δ ppm ): 11.35 (1H, s, NH), 10.97 (1H, s, NH), 7.62-6.84

(6H, m, Ar-H), 5.47 (1H, s, CH), 1.61 (2H, d, J = 6.6 Hz, CH3); 13C NMR (DMSO-

d6, δ ppm ): 164.6, 162.7, 157.6, 143.0, 132.5, 126.3, 126.2, 117.2, 116.6, 65.9

(CH2); EIMS (m/z, %): 457 (51, M+, Br81), 445 (33, M+, Br79), 422 (68, Br81), 420

(52, Br79), 342 (7), 296 (41, Br81), 294 (53, Br79), 268 (97, Br81), 266 (100, Br79),

240 (89, Br81), 242 (89, Br79), 191 (58, Br81), 189 (95, Br79), 162 (29), 145 (27),

103 (27), 75 (14), 63 (9); Anal. Cald for C17H12BrCl2N3O3: C, 44.67; H, 2.65; N,

9.19; Found: C, 44.66; H, 2.61; N, 9.15.

(E)-N′-(5-Bromo-2-oxoindolin-3-ylidene)-2-(2,4-dichlorophenoxy)butyrato-

hydrazide (92o): Yield: 86%; m.p: 207-208oC; IR (KBr, νmax, cm-1): 3547-3410

(NH stretching), 1636 (C=O), 1608 (C=N), 1561 (C=C), 1248 (C-O); 1H NMR

(DMSO-d 6, δ ppm ): 11.39 (1H, s, NH), 10.98 (1H, s, NH), 7.60-6.85 (6H, m, Ar-

H), 5.32 (1H, s, CH2), 2.88 (1H, s, CH2), 2.72 (1H, s, CH2); 13C NMR (DMSO-d 6,

δ ppm ): 164.6, 162.7, 157.6, 143.0, 132.5, 126.3, 126.2, 117.2, 116.6, 65.9

(CH2); EIMS (m/z, %): 473 (3, M+, Br81), 471 (3, M+, Br79), 445 (1, Br81), 443 (4,

Br79), 388 (1, Br81), 386 (2, Br79), 310 (98, Br81), 308 (100, Br79), 232 (9, Br81),

230 (33, Br79), 212 (8, Br81), 210 (10, Br79), 162 (12), 103 (7), 69 (13), 63 (3);

Anal. Cald for C18H14BrCl2N3O3: C, 45.89; H, 3.00; N, 8.92; Found: C, 45.82; H,

3.02; N, 8.95.

Page 297: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

293

Chapter 8

BIOLOGICAL 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 acts as a

tool to conduct activity directed isolations of bioactive compounds for curing

humans and animals. Primary screening provides first indication for bioactivities

and thus helps in the selection of lead compounds for secondary screening for

detailed pharmacological evaluations.

Indolinones, Triazoles and thiadiazoles were tested for the following

activities.

8.1. Herbicide studies

8.2. Fungicide studies

8.3. Insecticide studies

8.4. Plant growth regulating studies

8.5. Cytotoxicity and Antiviral activities against different cell culture

a. Vero cell culture

b. HeLa cell Culture

c. HEL cell Culture

d. Anti-feline corona virus in CRFK cell cultures

e. Anti-feline herpes virus in CRFK cell cultures

f. Anti-influenza virus activity in MDCK cell cultures

8.6. Antifungal studies

8.7. Antibacterial studies

The detail of each bioassay screening is as follows.

Page 298: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

294

8.1 Herbicide studies 8.1.1 Plant Material

Two broadleaf plants and two grasses were used to test the herbicidal

activity of compounds including rape (Brassica napus) (RA), amaranth pigweed

(Amaranthus retroflexus) (AR), barnyard grass (Echinochloa crusgalli (L.)

Beauv.) (BG) and Crab grass (Digitaria adscendens) (CG).

Seeds of amaranth pigweed, barnyard grass and crab grass were

reproduced outdoors and stored at 4°C. Seeds of rape were bought from Institute

of Crop, Tianjin Agroculture Science Academy.

8.1.2 Culture method

Seeds were planted in 7.5-cm-diameter disposable paper cup (250ml)

containing artificial mixed soil. Before plant emergence, the cups were covered

with plastic film to keep moist. Plants were grown in the green house. Fresh

weight of upground plants were measured 21 days after treatment.

8.1.3 Treatment

Dosage (activity ingredient) for each compound is 1500 grams per

hectare. Purified compounds were dissolved in 100µl N.N-dimethylformamide

with the addition of a little Tween 20, and then were sprayed using a laboratory

belt sprayer delivering at 750L/ha-spray-volume. The same amount of water was

sprayed as control.

a) Pre-emergency treatment: Compounds were sprayed immediately after

seeds planting. Two replicates each treatment.

b) Post-emergency treatment: Compounds were sprayed after the first true

leave expanding.

Page 299: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

295

Table 8.1a: Herbicide activity (% inhibition) of the synthesized

Chloroisatin derivatives 91(a-o)

BG RA AR

CG

Compd.

Density g / hect

ST FS ST FS ST FS ST FS 91a 1500 - - - - - - - - 91b 1500 - - - - - - - - 91c 1500 - - 2.4 22.3 22.5 13.9 - - 91d 1500 - - - - - - - - 91e 1500 - - - - - - - - 91f 1500 - - - - - - - - 91g 1500 - - 2.9 - - 16.5 - - 91h 1500 - - - - - - - - 91i 1500 - - 2.4 - - - - - 91j 1500 - - 36.0 30.4 22.5 8.9 - - 91k 1500 - - 7.8 34.9 - - - - 91l 1500 - - - - - 38.2 - -

91m 1500 - - 5.1 - - - - - 91n 1500 - - - - - - - - 91o 1500 - - 5.1 - - - - -

Table 8.1b: Herbicide activity (% inhibition) of the synthesized

Bromoisatin derivatives 92(a-o)

BG RA AR

CG

Compd.

Density g / hect

ST FS ST FS ST FS ST FS 92a 1500 - - - - - - - - 92b 1500 - - - - - - - - 92c 1500 - - - - - 13.9 - - 92d 1500 - - 20.0 - - - - - 92e 1500 - - - - - - - - 92f 1500 - - - - - 34.9 - 44.3 92g 1500 - - 33.3 - - - - - 92h 1500 - - - - - - - - 92i 1500 - - - - - 21.5 - - 92j 1500 - - 29.3 - - - - - 92k 1500 - - - - - - - - 92l 1500 - - - - - 24.5 - 45.2

92m 1500 - - - - - - - - 92n 1500 - - 59.1 - - - - - 92o 1500 - - - - - - - -

Key: (-) - No activity

Page 300: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

296

8.1.4 Analysis

The inhibition percent of upground fresh weight is used to describe the

control efficiency of compounds. Activity level:

A: ≥80%, B: 60~79%, C: 40~59%, D: ≤39%.

8.1.5 Discussion

From the biological assay results in Table 8.1a & 8.1b, which summarize

the herbicide activity of the synthesized compounds. From the results, it is infer

that a few compounds show herbicidal activity but not so significant.

8.2 Fungicide Studies 8.2.3 Disc method in vitro (DIV)

Disc method also called Agar dilution method. Including five kinds of fungi:

Fusarium wilt on cucumber (Fusarium oxysporum f. cucumerinum) (CF),

Speckle on peanut (Cercospora rachidicola) (PS), Tomato early blight

(Alternaria solani) (TB), Wheat scab (Gibberella zeae) (WS) and Apple

rootspot (Physalospora piricola) (AR).

8.2.2 Test concentration 50µg/ml. Detect mycelium expanding diameter 48h after treatment.

8.2.3 Against cucumber grey mould----Micro method in

vitro (MIV)

Spore suspension of cucumber grey mould (Botrytis cinerea) (CM)

contacted with compound solution to detect the inhibition activity of

compound to spore germination and growth of mycelium. Detect the result

by eyeballing 3 days after treatment.

Page 301: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

297

8.2.4 Against wheat powdery mildew----In vivo (IV)

Summer spores of wheat powdery mildew (Erysiphe graminis f. sp.

Tritici) (WP) were sprayed on wheat plant (3-leaf) and 24h later

compound suspension was sprayed as well. Disease index was

detected by eyeballing 7days after treatment.

8.2.5 Activity level

A: ≥80% B: 60~79%

C: 40~59% D: ≤39%

Table 8.2a: Fungicidal activity (% inhibition) of the synthesized

Chloroisatin derivatives 91(a-o)

DIV MIV IV Compd. Conc. (µg/ml) WS TB PS AR CF CM WP

91a 50 8.3 6.2 - 3.3 7.1 - - 91b 50 5.6 6.2 16.0 6.7 - - - 91c 50 - 3.1 - - 14.2 - - 91d 50 2.5 - - - - - - 91e 50 13.5 2.3 - 3.5 39.2 - - 91f 50 33.2 - - - - - - 91g 50 - 9.3 12.9 5.1 10.8 - - 91h 50 12.1 - 13.5 0 7.1 - - 91i 50 5.6 12.4 - 54.9 - - - 91j 50 15.4 - 4.0 5.1 - - - 91k 50 - 9.3 - 16.5 - - - 91l 50 - 18.6 - - - - -

91m 50 8.3 - 16.0 - - - - 91n 50 22.3 - 16.0 - - - - 91o 50 - - - 5.1 7.1 - -

Key: (-) - No activity

Page 302: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

298

Table 8.2b: Fungicidal activity (% inhibition) of the synthesized

Bromoisatin derivatives 92(a-o)

DIV MIV IV Compd. Conc. (µg/ml) WS TB PS AR CF CM WP

92a 50 8.3 6.2 - 3.3 7.1 - - 92b 50 5.6 6.2 16.0 6.7 - - - 92c 50 - 3.1 - - 14.2 - - 92d 50 2.5 - - - - - - 92e 50 13.5 2.3 - 3.5 39.2 - - 92f 50 33.2 - - - - - - 92g 50 - 9.3 12.9 5.1 10.8 - - 92h 50 12.1 - 13.5 - 7.1 - - 92i 50 5.6 12.4 - 54.9 - - - 92j 50 15.4 - 4.0 5.1 - - - 92k 50 - 9.3 - 16.5 - - - 92l 50 - 18.6 - - - - -

92m 50 8.3 - 16.0 - - - - 92n 50 22.3 - 16.0 - - - - 92o 50 - - - 5.1 7.1 - -

Key: (-) - No activity

8.2.6 Discussion

From the biological assay results in Table 8.2a and 8.2b, which

summarize the Fungicidal activity of the synthesized compounds. From the

results, it is inferred that a few compounds show fungicidal activity but not so

significant.

8.3 Insecticide Studies 8.3.1 Activity against Armyworm

Armyworm (Mythimna separata Walker)(AW),normal population feeding

indoor.

Page 303: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

299

Leaf- dipped-method: Dip maize leaf blade into compound solution (200 µg/ml,

dissolved in acetone), feeding 4th worm. Mainly observing stomach poison

activity, contact poison activity and feeding condition as well. Motility of worm

was detected 24h after treatment.

8.3.2 Activity against Aphid

Aphid (Aphis laburni kaltenbch)(AL), normal population feeding indoor on

bean plant.

Dipping-method: Dip plant with nymphae into compound emulsion (200µg/ml,

containing acetone or other solvent and Sorpol-560 or other emulsifier) for 2 to 3

seconds, swung off the liquid drop and then insert into the foam base covered

with glass mantle. Mainly observing stomach poison activity, contact poison

activity and feeding condition as well. Motility of worm was detected 24h after

treatment.

8.3.3 Activity level

A: ≥80% B: 60~79%

C: 40~59% D: ≤39%

8.3.4 Discussion

Insecticidal activity of the synthesized compounds is summarized in

Table 8.3. From the results, it is inferred that only compound 91b show very little

insecticidal activity (10%) and remaining chloro- and bromoisatin derivatives

show no insecticidal activity.

Page 304: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

300

Table 8.3: Insecticidal activity of the synthesized

Chloroisatin derivatives 91(a-o)

8.4 Plant growth regulating activity (PGR)

Modification of plant growth and development through the use of plant

growth regulators is becoming an increasingly important aspect of modern

agricultural practice. The availability of synthetic regulators that mimic the effect

of plant hormones has greatly facilitated this practice. Synthetic analogues of the

naturally occurring auxins, cytokinins and ethylene have been particularly useful

in this regard. However, most of the known plant growth regulators have

comparatively low physiological activity. The increase of the doses applied is

undesirable because of ecological reasons. All this imposes the search of new

high physiologically active substances.

Compd. Conc. (µg/ml) AW AL

91a 200 0 0 91b 200 10.0 0 91c 200 0 0 91d 200 0 0 91e 200 0 0 91f 200 0 0 91g 200 0 0 91h 200 0 0 91i 200 0 0 91j 200 0 0 91k 200 0 0 91l 200 0 0

91m 200 0 0 91n 200 0 0 91o 200 0 0

Page 305: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

301

8.4.1 Cucumber cotyledon rhizogenesis method (CCR) Filter paper method: To detect auxin-like activity of compound.

Concentration: 10µg.ml-1.

Cucumber cultivar: JINYAN 4.

Cotyledons were prepared by cultured on agar for 3 days (26±1°C) in dark

after seed immersion. 0.3ml 100µg.ml-1 solution dissolved in DMF was dropped

on paper disc with diameter of 6cm, air dried and put into Petri dishes (6cm). 3ml

distilled water was added into the dishes and then 10 cotyledons was put into as

well. Counting the number of adventitious roots arised and calculate the

increasing percent of rhizogenesis 5days after treatment (26±1°C).

8.4.2 Activity level A: ≥80% B: 60~79%

C: 40~59% D: ≤39%

Table 8.4: Plant growth regulating activity of the synthesized

Chloroisatin derivatives 91(a-o) and Bromoisatin derivatives 92(a-o)

Chloro-isatin derivatives Bromo-isatin derivatives

Compd. Cucumber cotyledon rhizogenesis method

(CCR) Compd.

Cucumber cotyledon rhizogenesis method

(CCR) 91a 23.3 92a 16.6 91b 46.6 92b 13.3 91c 70.0 92c 63.3 91d 0 92d 6.6 91e 20.0 92e 63.3 91f -30.0 92f -10.0 91g 12.5 92g -10.0 91h 61.7 92h 23.3 91i 54.1 92i 79.2 91j 75.1 92j 24.2 91k 73.0 92k -20.0 91l 16.6 92l 0

91m 13.3 92m 51.9 91n 40.0 92n 0 91o -20.0 92o -100.0

Page 306: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

302

8.4.3 Discussion

Plant growth regulatory activity of the synthesized compounds has been

summarized in Table 8.4. From the results, it is inferred that compounds 91c, 91j, 91k and 92i show significant activity while the remaining compounds also

show activity but not so significant.

8.5 Antiviral Studies (For experimental detail, sec section 4.9 of this thesis)

Table 8.5a: Cytotoxicity and Antiviral activities of Chloroisatin derivatives

91(a-o) in HeLa cell Culture

EC50b (µg/ml)

Compound Minimum cytotoxic

concentrationa (µg/ml)

Vesicular stomatitis

virus Coxsackie virus B4

Respiratory syncytial

virus 91a 100 >20 >20 >20 91b ≥20 >20 >20 >20 91c 20 >4 >4 >4 91d 100 >20 >20 >20 91e 100 >20 >20 >20 91f 20 >4 >4 >4 91g 20 >4 >4 >4 91h 20 >4 >4 >4 91i 4 >0.8 >0.8 >0.8 91j ≥20 >20 >20 >20 91k 100 >20 >20 >20 91l 20 >4 >4 >4

91m 20 >4 >4 >4 91n 20 >4 >4 >4 91o ≥20 >20 >20 >20

D.S.-5000 >100 2.4 12 0.5 (S)-DHPA (µM) >250 30 >250 150 Ribavirin (µM) >250 10 250 10

Page 307: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

303

Table 8.5b: Cytotoxicity and Antiviral activities of Bromoisatin derivatives

92(a-o) in HeLa cell Culture

EC50b (µg/ml)

Compound Minimum cytotoxic

concentrationa (µg/ml)

Vesicular stomatitis

virus Coxsackie virus B4

Respiratory syncytial

virus 92a 20 >4 >4 >4 92b 100 >20 >20 >20 92c 0.8 >0.16 >0.16 >0.16 92d 100 >20 >20 >20 92e 20 >4 >4 >4 92f ≥20 20 >20 >20 92g 20 >4 >4 >4 92h 100 >20 >20 >20 92i >100 >100 >100 >100 92j 20 >4 >4 >4 92k 20 >4 >4 >4 92l 20 >4 >4 >4

92m 100 >20 >20 >20 92n ≥20 >20 >20 >20 92o 20 >4 >4 >4

D.S.-5000 >100 2.4 12 0.5 (S)-DHPA (µM) >250 30 >250 150 Ribavirin (µM) >250 10 250 10

Page 308: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

304

Table 8.5c: Cytotoxicity and Antiviral activities of Chloroisatin derivatives

91(a-o) and Bromoisatin derivatives 92(a-o) in Vero cell cultures

EC50

b (µg/ml)

Compound Minimum cytotoxic

concentrationa (µg/ml)

Para-influenza-

3 virus

Reovirus-1

Sindbis virus

Coxsackievirus

B4

Punta Toro virus

91a >100 >100 >100 >100 >100 >100 91b 100 >20 >20 >20 >20 >20 91c 20 >4 >4 >4 >4 >4 91d 100 >20 >20 >20 >20 >20 91e 100 >20 >20 >20 >20 >20 91f ≥100 >100 >100 >100 60 >100 91g ≥0.8 >0.8 >0.8 >0.8 >0.8 >0.8 91h 20 >4 >4 >4 >4 >4 91i 20 >4 >4 >4 >4 >4 91j 20 >4 >4 >4 >4 >4 91k 100 >20 >20 >20 >20 >20 91l ≥20 >20 >20 >20 >20 >20

91m 20 >4 >4 >4 >4 >4 91n ≥20 >20 >20 >20 >20 >20 91o 100 >20 >20 >20 >20 >20 92a 100 >20 >20 >20 >20 >20 92b 20 >4 >4 >4 >4 >4 92c ≥100 >100 >100 >100 >100 60 92d 20 >4 >4 >4 >4 >4 92e 20 >4 >4 >4 >4 >4 92f >100 >100 >100 >100 >100 >100 92g ≥20 >20 >20 >20 >20 >20 92h 100 >20 >20 >20 >20 >20 92i ≥100 >100 >100 >100 >100 >100 92j 20 >4 >4 >4 >4 >4 92k 20 >4 >4 >4 >4 >4 92l ≥4 >4 >4 >4 >4 >4

92m >100 >100 >100 >100 >100 >100 92n 100 >20 >20 >20 >20 >20 92o 100 >20 >20 >20 >20 >20

D.S-5000 >100 >100 >100 20 12 >100 (S)-DHPA (µM) >250 >250 >250 >250 >250 >250 Ribavirin (µM) >250 150 250 250 >250 150

Page 309: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

305

Table 8.5d: Cytotoxicity and Antiviral activities of Chloroisatin derivatives 91(a-o) and Bromoisatin derivatives 92(a-o) in HEL cell Culture

EC50b (µg/ml)

Compound Minimum cytotoxic

concentrationa (µg/ml)

Herpes simplex virus-1 (KOS)

Herpes simplex virus-2

(G)

Vaccinia virus

Vesicular stomatitis

virus

Herpes simplex virus-1

TK- KOS ACVr

91a >100 >100 >100 >100 >100 >100 91b 100 >20 >20 >20 >20 >20 91c 20 >4 >4 >4 >4 >4 91d 100 >20 >20 >20 >20 >20 91e 100 >20 >20 20 >20 >20 91f 100 >20 >20 >20 >20 >20 91g 0.8 >0.16 >0.16 >0.16 >0.16 >0.16 91h 20 >4 >4 >4 >4 >4 91i 20 >4 >4 >4 >4 >4 91j 100 >20 >20 >20 >20 >20 91k 100 >20 >20 >20 >20 >20 91l ≥4 >4 >4 >4 >4 >4

91m 0.8 >0.16 >0.16 >0.16 >0.16 >0.16 91n 100 >20 >20 >20 >20 >20 91o 100 >20 >20 >20 >20 >20 92a 100 >20 >20 >20 >20 >20 92b 100 >20 >20 >20 >20 >20 92c 100 >20 >20 >20 >20 >20 92d 100 >20 >20 20 >20 >20 92e 20 >4 >4 >4 >4 >4 92f 100 >20 >20 >20 >20 >20 92g 20 >4 >4 >4 >4 >4 92h 20 >4 >4 4 >4 >4 92i ≥20 >20 >20 >20 >20 >20 92j 4 >0.8 >0.8 >0.8 >0.8 >0.8 92k ≥0.8 >0.8 >0.8 >0.8 >0.8 >0.8 92l ≥0.8 >0.8 >0.8 >0.8 >0.8 >0.8

92m 100 >20 >20 >20 >20 >20 92n 100 >20 >20 >20 >20 >20 92o 100 >20 >20 >20 >20 >20

Brivudin (µM) >250 0.08 10 6 >250 50 Ribavirin (µM) >250 10 150 250 >250 150 Acyclovir (µM) >250 0.4 0.4 250 >250 50

Ganciclovir (µM) >100 0.03 0.03 100 >100 2 aRequired to cause a microscopically detectable alteration of normal cell morphology. bRequired to reduce virus-induced cytopathogenicity by 50 %.

Page 310: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

306

Table 8.5e: Cytotoxicity and anti-Feline Corona Virus (FIPV) and anti-Feline

Herpes Virus activity of chloroisatin derivatives 91(a-o) and Bromoisatin

derivatives 92(a-o) in CRFK cell cultures EC50

b (µg/ml)

Compounds CC50a (µg/ml) Feline Corona Virus

(FIPV) Feline Herpes Virus

91a >100 46.7 >100 91b >100 >100 >100 91c 12.9 >4 >4 91d 31.9 >20 >20 91e >100 >100 >100 91f >100 >100 >100 91g >100 >100 >100 91h 5.7 >4 >4 91i 17.6 >4 >4 91j 24.4 >20 >20 91k >100 >100 >100 91l >100 >100 >100

91m >100 >100 >100 91n >100 >100 >100 91o >100 >100 >100 92a >100 >100 81.2 92b >100 >100 >100 92c 42.5 >20 >20 92d 22.9 >20 >20 92e 11.8 >4 >4 92f >100 >100 >100 92g 96.7 >20 >20 92h 69.0 >20 >20 92i >100 >100 >100 92j 2.6 >0.8 >0.8 92k >100 >100 >100 92l >100 >100 >100

92m 5.7 >4 >4 92n >100 >100 >100 92o >100 >100 >100 HHA >100 3.2 6.4 UDA >100 6.6 5.2

Ganciclovir (µM) >100 >100 2.6 a 50% Cytotoxic concentration, as determined by measuring the cell viability with the colorimetric formazan-based MTS assay. b 50% Effective concentration, or concentration producing 50% inhibition of virus-induced cytopathic effect, as determined by measuring the cell viability with the colorimetric formazan-based MTS assay. CRFK cells: Crandell-Rees Feline Kidney cell

Page 311: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

307

8.5f Anti-influenza virus activity and cytotoxicity in MDCK cell cultures 8.5.1 Procedure

The test compounds were evaluated for their antiviral activity against three

influenza virus subtypes [A/Puerto Rico/8/34(H1N1); A/Hong Kong/7/87 (H3N2)

and B/Hong Kong/5/72]. Antiviral activity was estimated from the inhibitory effect

on virus-induced cytopathic effect, as determined by microscopical examination

and/or the formazan-based MTS cell viability test. Cytotoxicity of the test

compounds was expressed as the compound concentration causing minimal

changes in cell morphology (MCC), or the concentration causing 50% cytotoxicity

(CC50), as determined by the MTS assay.

8.5.2 Discussion

Compounds 91(a-o) and 92(a-o) were evaluated for their anti-influenza

virus activity, and their activities were compared with those of oseltamivir

carboxylate (the active form of Tamiflu®), ribavirin, amantadine and rimantadin

(Table 8.5f). None of the compounds 91(a-o) and 92(a-o) were able to inhibit the

cytopathic effects of influenza A or B at subtoxic concentrations. On the contrary,

the reference compounds oseltamivir carboxylate and ribavirin were active

against influenza virus; their EC50 values are clearly lower than their MCC values

(concentrations causing minimal toxicity). For amantadine and rimantadine, the

best activity was seen with the H3N2 strain. These compounds are known to be

inactive against influenza B. Also, the H1N1 A/PR/8/34 strain that is used in our

tests is less sensitive to amantadine and rimantadine.

Page 312: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

308

Table 8.5f: Cytotoxicity and Anti-influenza acticity of chloroisatin derivatives 91(a-o)

Cytotoxicity Antiviral EC50

c Influenza A

H1N1 subtype

Influenza A H3N2

subtype Influenza B

Compd. Conc. unit

Minimum cytotoxic

conc.a CC50

bvisualCOE score

MTSvisual CPE

score MTS

visualCPE

score MTS

91a µg/ml 100 >100 NA NA NA NA NA NA 91b µg/ml 20 >100 NA NA NA NA NA NA 91c µg/ml 20 30.3 NA NA NA NA NA NA 91d µg/ml 20 11.3 NA NA NA NA NA NA 91e µg/ml 4 2.0 NA NA NA NA NA NA 91f µg/ml ≥100 >100 NA NA NA NA NA NA 91g µg/ml ≥4 21.5 NA NA NA NA NA NA 91h µg/ml ≥20 42.9 NA NA NA NA NA NA 91i µg/ml 100 >100 NA NA NA NA NA NA 91j µg/ml 20 9.0 NA NA NA NA NA NA 91k µg/ml 20 100 NA NA NA NA NA NA 91l µg/ml 4 1.9 NA NA NA NA NA NA

91m µg/ml 100 >100 NA NA NA NA NA NA 91n µg/ml 100 >100 NA NA NA NA NA NA 91o µg/ml 4 2 NA NA NA NA NA NA

Oseltamivir carboxylate µM >100 >100 0.07 0.08 107 1.5 4 2.0

Ribavirin µM 100 94.6 9 12.4 9 8.2 9 5.5 Amantadin µM >100 >100 45 83.3 4 3.1 NA NA Rimantadin µM >100 >100 45 39.3 0.8 0.1 NA NA

Page 313: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

309

Table 8.6g: Cytotoxicity and Anti-influenza acticity of Bromoisatin derivatives 92(a-o)

Cytotoxicity Antiviral EC50c

Influenza A H1N1

subtype

Influenza A H3N2

subtype Influenza B

Compd. Conc. unit

Minimum cytotoxic

conc.a CC50

bvisualCOE score

MTSvisual CPE

score MTS

visualCPE

score MTS

92a µg/ml 100 >100 NA NA NA NA NA NA 92b µg/ml 20 >100 NA NA NA NA NA NA 92c µg/ml 20 30.3 NA NA NA NA NA NA 92d µg/ml 20 11.3 NA NA NA NA NA NA 92e µg/ml 4 2.0 NA NA NA NA NA NA 92f µg/ml ≥100 >100 NA NA NA NA NA NA 92g µg/ml ≥4 21.5 NA NA NA NA NA NA 92h µg/ml ≥20 42.9 NA NA NA NA NA NA 92i µg/ml 100 >100 NA NA NA NA NA NA 92j µg/ml 20 9.0 NA NA NA NA NA NA 92k µg/ml 20 100 NA NA NA NA NA NA 92l µg/ml 4 1.9 NA NA NA NA NA NA

92m µg/ml 100 >100 NA NA NA NA NA NA 92n µg/ml 100 >100 NA NA NA NA NA NA 92o µg/ml 4 2 NA NA NA NA NA NA

Oseltamivir carboxylate µM >100 >100 0.07 0.08 107 1.5 4 2.0

Ribavirin µM 100 94.6 9 12.4 9 8.2 9 5.5 Amantadin µM >100 >100 45 83.3 4 3.1 NA NA Rimantadin µM >100 >100 45 39.3 0.8 0.1 NA NA

b50% Cytotoxic concentration, as determined by measuring the cell viability with the colorimetric formazan-based MTS assay. aMinimum compound concentration that causes a microscopically detectable alteration of normal cell morphology. c50% Effective concentration, or concentration producing 50% inhibition of virus-induced cytopathic effect, as determined by visual scoring of the CPE, or by measuring the cell viability with the colorimetric formazan-based MTS assay. MDCK cells: Madin Darby canine kidney cells. NA: not active at the highest concentration tested, or at subtoxic concentration

Page 314: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

310

8.5.3 Conclusion

The reference compounds oseltamivir carboxylate (the active form of

Tamiflu®) and ribavirin were active against influenza virus; their EC50 values are

clearly lower than their MCC values (concentrations causing minimal toxicity).

For amantadin and rimantadin, the best activity was seen with the H3N2 strain

(middle column). These compounds are known to be inactive against influenza B

(right column). Also, the H1N1 A/PR/8/34 strain that is used in our tests is known

to be less sensitive to amantadine and rimantadine (left column).

Among the range of 30 compounds [chloroisatin 91(a-o) and bromoisatin

92(a-o)] tested, none was able to inhibit the cytopathic effects of influenza A or B

virus at subtoxic concentrations or the highest concentration tested (100 µg/ml).

8.6 Antifungal Studies During last two decades, the life threatening infections caused by

pathogenic fungi and bacteria become increasingly common, especially in

individuals immunocompromised patients with AIDS. Clinically, candidosis,

aspergillosis, and cryptococosis are major fungal infections in these patients.

Fungi also produced toxin in foods and cause poisoning with out being physically

present. Another problem caused by fungi is allergy due to their spores.

However, the current antifungal therapy suffers from drug related toxicity; sever

drug resistance, non optimal pharmacokinetics and serious drug interaction.

Therefore, there is an emergent need to develop novel antifungal drugs with

higher efficiency, broad spectrum and low toxicity. Therefore antifungal and

antibacterial activities of synthesized triazoles 89(a-t) and thiadiazoles 90(a-j) were carried out.

Page 315: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

311

8.6.1 Antifungal activities of synthesized triazole 89(a-t) and thiadiazoles 90(a-j)

The agar tube dilution method was used for testifying the antifungal

activities of synthesized triazoles and thiadiazoles. The antifungal assay was

done against four different fungal strains, which are:

Aspergillus flavus

Mucor species

Aspergillus niger

Aspergillus fumigatus

These funguses were maintained on sabouraud dextrose agar (SDA)

medium at 4ºC.

8.6.2 Media for fungus Sabouraud dextrose agar (SDA) was used to grow fungus for inoculums

preparations. Its composition was:

Peptone complex = 10gm/L

Glucose = 40 gm/L

Agar =15 gm/L

8.6.3 Preparation of media for fungus Sabouraud dextrose agar (SDA) was prepared by dissolving 6.5 gm

/100mL in distilled water and PH was adjusted at 5.6. Contents were dissolved

and dispensed as 4 mL volume into screw capped tubes and were autoclaved at

121 ºC for 21 minutes.

Page 316: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

312

8.6.4 Loading of samples Tubes were allowed to cool to 50ºC and non solidified SDA was loaded

66.6 µL of triazoles 89(a-t) and thiadizoles 90(a-j) with pipette from stocked

solution. This would give the final concentration of 200 µg/ml of the pure

compound in the media. Tubes were then allowed to solidify at room temperature

in slanting position. Tubes were prepared in triplicate for each fungus species. 8.6.5 Inoculation of fungus, incubation and measurement

of growth inhibition The tubes containing solidified media and tested triazoles 89(a-t) and

thiadizoles 90(a-j) were inoculated with 4mm diameter of inoculums, taken from

seven days old culture of fungus. Other media supplemented with DMSO and

terbinafine were used as negative and positive control respectively. All

experiments were done in three replicates. The tubes were incubated at 28ºC for

seven days. Growth in the media was determined by measuring linear growth

(cm) and growth inhibition was calculated with reference to negative control.

(cm) control negativein growth Linear m)in test/(cgrowth linear - (cm) control negativein growth Linear )Inhibition (%age Formula =

8.6.6 Activity level

Below 40% inhibition = low activity

40-60% inhibition = moderate activity

60-70% inhibition = good activity

70% inhibition = significant activity

Page 317: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

313

Table 8.6a: Antifungal Assay of Triazoles 89(a-t)

% inhibition Compd. Aspergillus

flavus Aspergillus fumigatus

Aspergillus niger

Mucor. sp

89a 36.58 32.86 74.89 45.45 89b 49.08 45.10 45.41 11.03 89c 16.61 10.56 19.58 17.20 89d 48.48 20.87 29.16 7.90 89e 56.71 12.5 40.20 1.08 89f 39.48 17.78 43.33 5.73 89g 25.76 47.03 37.50 17.64 89h 9.60 15.97 12.91 13.41 89i 4.72 30.79 20.62 40.25 89j 18.59 20.10 8.0 18.72 89k 34.91 41.10 52.50 21.86 89l 46.79 29.63 66.56 10.28

89m 57.47 42.91 34.47 4.65 89n 40.20 39.48 1.08 49.08 89o 43.33 25.76 5.73 16.61 89p 37.50 9.60 17.64 48.48 89q 12.91 4.72 13.41 56.71 89r 20.62 34.91 40.25 39.48 89s 29.63 46.79 17.64 25.76 89t 42.91 57.31 13.41 20.10

Turbenafine 100 100 100 100

Table 8.6: Antifungal Assay of Thiadizoles 90(a-j)

% inhibition Compd. Aspergillus

flavus Aspergillus fumigatus

Aspergillus niger

Mucor. sp

89a 22.05 52.24 49.47 51.58 89b 25.91 51.82 50.10 51.79 89c 30.12 44.57 49.68 51.58 89d 8.67 22.16 29.17 30.65 89e 14.45 13.49 15.01 15.01 89f 10.91 12.15 38.99 44.95 89g 16.38 19.75 23.890 42.91 89h 13.91 13.91 11.62 14.58 89i 16.38 19.75 23.89 42.91 89j 12.77 44.09 50.52 52.43

Turbenafine 100 100 100 100

Page 318: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

314

8.6.6 Interpretation of results

Synthesized triazoles 89(a-t) and thiadizoles 90(a-j) were screened for

their antifungal activities. In vitro evaluation of antifungal activity was carried out

by the agar tube dilution method. All triazoles and thiadizoles exhibited moderate

activity against four fungal strains compared to reference chemotherapeutic i.e.

terbinafine at tested concentration. Among these synthesized compounds, 89a

and 89l were most active against Aspergillus niger. All the other remaining

compounds also showed good to moderate activity against these four fungal

strains.

8.7 Antibacterial assay

Synthesized triazoles 89(a-t) and thiadizoles 90(a-j) were screened at 1

mg/mL in DMSO (initial concentration of compounds). Nutrient broth medium for

bacterial growth was prepared by dissolving 0.8g/100mL in distilled water & pH

was adjusted to 7.2 and then medium was autoclaved. To performed

antibacterial assay, Nutrient agar medium was prepared by dissolving 2g/100mL

in distilled water and PH was maintained at 7.2, and then medium was

autoclaved. To compare the turbidity of bacterial culture, McFarland 0.5 barium

sulphate was used.

Following bacterial strains were used:

Staphylococcus aurens (Gram positive)

Bacillus Subtilis (Gram positive)

Salmonella setubal (Gram negative)

Enterobacter aerogenes (Gram negative)

Page 319: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

315

8.7.1.1 Method

Nutrient agar medium was prepared by above mention method. It was

sealed with 1mL of prepared oculum/100mL of prepared nutrient agar medium

and was shaked. Petri plates (14cm) were made by pouring 75mL of sealed

nutrient agar media and allow it to solidify. Eleven wells per plate were made with

sterile cork borer (8mm). Using micropipette, 100µL of test solutions were poured

in respective wells. To all samples two solutions for positive control

(Roxithromycin 1mg/mL, Cefixime-USP 1mg/mL and one negative control DMSO

was applied to each plate. These plates were incubated at 37ºC. After 24 hour

incubation diameter of the clear zone was measured.

8.7.2 Results These synthesized triazoles 89(a-t) and thiadizoles 90(a-j) show no

activity against above mentioned bacteria.

Page 320: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

133

REFERENCES (PART ONE)

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. Cousse, H.; Mouzin, G.; Fr. Demande, 1975, 2, 317922

(C1.A61k31/37).

14. Chinworrungsee, M.; Kittakoop, P.; Isaka, M.; Chanphen, R.;

Tanticharoen, M.; Thebtaranonth, Y. J. Chem. Soc., Perkin Trans. 1,

2002, 2473.

15. Mizushima, M.; Oki, S. Hapan Kokai., 1977, JP 77, 83, 374 (C1. C07D

311/76).

16. Blum, M. S.; Jones, T. H.; Howard, D. F.; Overal, W. L. Comp. Biochem.

Physiol., 1982, 71B, 731.

17. Brophy, J. J.; Cavil, G. W. K.; Plant, W. D. Insect Biochem., 1981, 11, 307.

Page 321: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

134

18. Brand, J. M.; Fales, H. M.; Sokosloski, E. A.; Macconell, J. G.; Duffield, R.

M. Life Science, 1980, 13, 2003.

19. Baker, T. C.; Nishida, R.; Roelofs, W. Science, 1981, 214, 1359.

20. Lloyd, H. A.; Evans, S. L.; Khan, A. H.; Tscinkel, W. R.; Blum, M. S. Insect

Biochem., 1978, 8, 336.

21. Waitz, J. A.; Drube, C. G. Antifungal Agents. Annu. Rcp. Med. Chem.,

1972, 7, 109.

22. Nakajima, S.; Kawai, K.; Yamada, S.; Sawai, Y. Agric. Biol. Chem., 1976, 40, 811.

23. Scott, P. M.; van Walbeek, W. J. Antibiotics, 1971, 24, 747.

24. Davies, W. P.; Lewis, B. G. New phytol., 1981, 89, 109.

25. Nakajima, S.; Sugiyama, S.; Suto, M. Org. Prep. Proced. Int., 1979, 11,

77.

26. Ceska, O.; Chaudhary, S. K.; Warrington, P.; Ashwood-Smith, M. J.;

Bushnell, G. W.; Poulton, G. A., Phytochem. Photobio. 1988, 27, 2083.

27. Hudson, J. B.; Graham, E. A.; Harris, L.; Ashwood-Smith, M. J. 1993, 57, 491.

28. Clark, M. T.; Gilmore, I. J. Uk Pat. Appl., 1989, GB 2207425, C.A. 111: 19479V.

29. Yoshikawa, H.; Taniguchi, E.; Meakawa, K. Nippon noyaku gakkaishi, 1979, 4, 475, Ibid. 1980, 5, 1.

30. Sanawa kagaku kenkysho Co. Ltd. Jpn. Kokai Tokyo koho, 1983, Jp 58,

162, 586[83,162. 586] (C1. C07D 311/76).

31. Gremaud, G.; Tabacchi, R. Nat. Product Letters, 1994, 5, 95.

32. Kameda, K.; Aoki, H. et al., Agric. Biol. Chem., 1973, 37, 2137.

33. Kumagai, H. et al., Journal of Antibiotics, 1990, 72, 1505.

34. Powers, J. C.; Harper, W.; U. S. P. 1984, US 4596, A22 (C1.514-459;

A61k31/35).

35. Okuno, T.; Oikawa, S.; Goto, T.; Sawai, K.; Shirahama, H.; Matsumoto, T.

Agic. Biol. Chem., 1986, 50, 997.

Page 322: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

135

36. Bihel, F.; Quelever, G.; Lelouard, H.; Petit, A.; Alves-de-Costa, C.;

Pourquie, O.; Checler, F.; Thellend, A.; Pierre, P.; Karaus, J. Bioorg Med

Chem. 2003,11, 3141.

37. Kang, S. Y.; Lee, K. Y.; Sung, S. H.; Park, M. J.; Kim, Y. C. J. Nat. Prod.

2001,64,683.

38. Thrash, T. P.; Welton, T. D.; Behar, V. Tetrahedron Lett. 2000, 41, 29.

39. Atanasova, I.; Khimaova, M.; Chaidarova, V.; Nakaov, A.; Petkov, N.;

Avramova, R. Jpn. Kokai Tokyo Koho., 1986, JP, 61, 238, 788 (86, 238,

788) (C1. C07D 499/68).

40. Yuan, H.; Junker, B.; Helquist, P.; Taylor, R. E. Current Organic Synthesis,

2004, 1, 1-9.

41. Bauta, W.E.; Lovett, D. P.; Cantrell, W. R.; Bruke, B.D. J. Org. Chem. 2003,

68, 5967-5973

42. Yin, L, Ohno T, Weichselbaum R, Kharbanda S, Kufe D. Mol. Cancer Ther.

2001; 1, 43– 48.

43. Nakashima, T., Hirano, S., Agata, N., Kumagai, H., Isshiki, K., Yoshioka, T.,

Ishizuka, M., Maeda, K., and Takeuchi, T. J. Antibiot. (Tokyo), 1999, 52,

426–428.

44. Bonate, P. L.; Eder, J. P.; Soulie, P. Proc Am Soc Clin Oncol. 2003, 22,

134.

45. Salloum, R. M.; Jaskowiak, N. T.; Mauceri, H. J.; Seetharam, S.; Beckett,

M. A.; Koons, A. M.; Hari, D. M.; Gupta, V. K.; Reimer, C.; Kalluri, R.;

Posner, M. C.; Hellman, S.; Kufe, D. W.; Weichselbaum, R. R. Cancer

Res. 2000, 60, 6958.

46. Naoki, A.; Hiroko, N.; Michael, M.; Surender, K.; Donald, K. Cancer Res.

2004, 64, 8512.

47. Corinne, L.; Reimer, N. A.; Jennifer, G.; Tammam, M. B.; William M. D.;

George, D. K.; Susan L. R.; Michael, M.; Robert, F.; Raghu, K.; Donald,

K.; Surender, K.; Cancer Res. 2002, 62, 789.

Page 323: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

136

48. Ozoe, Y.; Kuriyama, T.; Tachibana, Y.; Harimaya, K.; Takahashi, N.;

Yaguchi, T.; Suzuki, E.; Imamura, K., Oyama, K. J. Pestic. Sci., 2004, 29,

328.

49. Eun-Jeon, P.; Hyuncheol, O.; Tai-Hyun, K.; Dong-Hwan, S.; Youn-Chul, K.

Arch Pharm Res. 2004, 27, 944.

50. Dae-Seog, L.; Yi-Sub, K.; Kyung-Ho, L.; Si-Hwan, K.; Won-Ho, Y.; Won-

Young, L.; Chang-Han, K. Chemotherapy , 2003, 49, 257.

51. Engelmeier, D.; Hadacek, F.; Hofer, D.; Kutchera, G. L.; Nagl, M.; Wurz,

G.; Greger, H. J. Nat. Prod. 2004, 67, 19.

52. Kam, C. M.; Fujikawa, K.; Powers, J. C. Biochemistry, 1988, 27, 2547.

53. Oweida, W. S.; Ku, N. D.; Lumsden, A. B.; Kam, C. M.; Powers, J. C.

Thromb. Res., 1990, 58, 191.

54. Kam, C. M.; Oglesby, T. J.; Pangburn, M. K.; Volanakis, J. E.; Powers, J. C.

J. Immunol., 1992, 149, 163.

55. Claydon, N.; Grove, J. F.; Hoshen, M. Chem. and Ind., 1974, 344.

56. Phenolic Metabolites of Caulo Ulmi, Phytochem., 1974, 13, 2567.

57. Kameda, K.; Aoki, H. Agric. Biol. Chem., 1973, 37, 2537.

58. Bestman, H. J.; Kern, F.; Schafe, D.; Witschel, M. C. Angrew. Chem. Int.

Ed. Eng., 1992, 31, 795.

59. Sassa, T.; Adki, G. Agric. Biol. Chem., 1968, 32, 1432.

60. Satomura, Y.; Sato, A. Agric. Biol. Chem., 1965, 29, 337.

61. Hemingway, R. W.; Mc Graw, H. W. J. Agric. Food Chem., 1977, 25, 717.

62. Houlchen, W. J.; Nadelson, J. British patent. 1975, 1374337 (1874); C.A.

83, 43196.

63. Piller, N. B. Lymphology, 1976, 9, 132.

64. Kuhr, I.; Fuska, J.; Sedmera, P. ; Podojil, M. ; Vokoun, J. ; Vanek, Z. J.

Antibiotics, 1973, 26, 535.

65. Munakata, T.; Okumoto, T. Chem.Pharm. Bull., (Japan), 1981, 29, 891.

66. Monakata, T.; Bactobolins, Yakugaku Zasshi, 1981, 101, 138 (1981); C.A.

95, 42826.

Page 324: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

137

67. Itoh, J.; Shomura, T.; Omoto, S.; Miyato, S.; Yuda, Y. ; Shibata, U.; Inouye,

S. Agric. Biol. Chem., 1982, 46, 1255.

68. Shimojima, Y.; Hayashi, H.; Ooka, T.; Shibkawa, M.; Ittaka, Y. Tetrahedron, 1984, 40, 2519.

69. Liu, D. Zhonghua Yixue Zashi, 1982, 62, 336.

70. Houlihan, W. J.; Nadelson, J. Fr. Demande, 1973, 2, 178, 718 (C1. A 61K

C07d).

71. Steyn, P. S. Microbial toxins, 1971, 6, 179.

72. Vleggaar, R.; Steyn, P. S. The biosynthesis of some miscellaneous

mycotoxins.; Academic Press, New Yark, 1980.

73. Muller, E.; Gawlick, H.; Kreutzman, W. Ann. Chem., 1934, 97, 515.

74. Umezawa, H. Pure Appl. Chem., 1973, 33, 129.

75. Furutani, Y.; Naganawa, H.; Takeuchi, T.; Umezawa, H. Agric. Biol Chem., 1977, 41, 1179.

76. Money, T.; Comer, F. W.; Webster, G. B.; Wright, I. G.; Scott, A. I.

Tetrahedron, 1972, 111, 740 (Ital.).

77. Dean, F. M. naturally occurring oxygen ring compounds, 1963,

Butterworths, London, pp.471.

78. Turner, W. B. Fungal metabolites, 1971, Academic Press London.

79. Suzuki, Y. Agric. Biol. Chem; (Japan), 1970, 34, 760.

80. Afzal, S. M.; Pikes, R.; Rama, N. H.; Smith, I. R.; Turner, P. S.; Whalley, W.

B. J. Chem. Soc; Perkin Transactions I., 1978, 81.

81. Feliciano, A. S.; Miguel del Corral, J. M.; Canedo, L. M.; Medrade, M. Phyto

Chem., 1990, 29, 945.

82. Gripeneberg, J. Acta Chem. Scan., 1971, 25, 2999.

83. Rama, N. H.; Saeed, A.; Bird, C. W. Liebigs Ann. Chem., 1993, 1331.

84. Rama, N. H.; Saeed, A.; Bird, C. W. Liebibg Ann. Chem., 1995, 711.

85. Billek, G. Kindl, H. Monatsh. Chem., 1962, 93, 85.

86. Billek, G.; Kindl, H. Monatsh. Chem., 1962, 93, 814.

87. Ibrahim, R. K.; Towers, G. H. N. Canad. J. Biochem. Physiol., 1962, 40,

449.

Page 325: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

138

88. Yagi, A.; Ogata, Y.; Yamauchi, T.; Nishioka I. Phytochemistry, 1977, 16,

1098.

89. Taneyama, M.; Yoshida, S. Botan. Mag., 1979, 92, 69.

90. Mali, R. S.; Jagtap, P. G.; Patil, S. R.; Pawar, P. N. J. Chem. Soc. Chem.

Commun., 1992, 883.

91. Narasimhan, N. S.; Mali, R. S. Topics in current chemistry, 1987, 138, 63.

92. Woon, E. C. Y.; Dhami, A.; Mahon, M. F.; Threadgill, M. D. Tetrahedron.

2006, 62, 4829.

93. Subramanian, V.; Batchu, V. R.; Barange, D.; Pal, M. J. Org. Chem. 2005,

70, 4778.

94. Roy, H.; Sarkar, M. Synth. Commun. 2005, 35, 2177.

95. Cherry, K.; Parrain, J. L.; Thibonnet, J.; Duchene, A.; Abarbri, M. J. Org.

Chem. 2005, 70, 6669.

96. Suzuki, T.; Yamada, T.; Watanabe, K.; Katoh, T. Bioorg. Med. Chem. Lett.

2005, 15, 2583.

97. Opatz, T.; Ferenc, D. Eur. J. Org. Chem. 2005, 817.

98. Martinez, A.; Fernandez, M.; Estevez, J. C.; Estevez, R. J.; Castedo, L.

Tetrahedron 2005, 61, 485.

99. Yao, T.; Larock, R. C. J. Org. Chem. 2003, 68, 5936.

100. Liao, H.-Y.; Cheng, C. H. J. Org. Chem. 1995, 60, 3711.

101. Hussain, M.; Rama, N. H.; Hameed, S.; Malik, A.; Khan, K. M. Nat.

Prod. Res. 2005, 19, 41.

102. Zamani, K.; Faghihi, K.; Ebrahimi, S. Turk. J. Chem. 2005, 29, 171.

103. Narasimhan, N.S.; Mali, R. S. Synthesis, 1983, 63.

104. Snieckus, V. Chem. Rev., 1990, 90, 879.

105. Lee, D.; Still, W.C. J. Org. Chem., 1989, 54, 4715.

106. Reitz, D. B.; Massey, S. M. J. Org. Chem., 1990, 55, 1375.

107. Superchi, S.; Minutolo, F.; Pini, D.; Salvadori, P. J. Org. Chem., 1996, 61,

3183.

108. Mroady, S. M.; Rexhausen, J. E.; Thomas, E. J. J. Chem. Soc. Perkin

Trans. 1., 1999, 1083.

Page 326: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

139

109. Pini, D.; Superchi, S.; Salvadori, P. J. Organometallic. Chem., 1993, 452.

110. Gruniwald, G. L.; Dahanukar, V. H. J. Heterocyclic Chem., 1994, 31, 1609.

111. Bestmann, H. J.; Kern, F.; Schafer, D.; Witschel, M. C. Angew. Chem., Int.

Ed. Engl., 1992, 31, 795.

112. Choukchou-Braham, N.; Asakawa, Y.; Lepoittevin, J. P. Tetrahedron Lett., 1994, 35, 3949.

113. Bhide, B. H.; Akolkar, V. D.; Brahmbhat, D. I. Ind. J. Chem., 1992, 31(B),

116.

114. Kurosaki, Y.; Fukuda, T.; Iwao, M. Tetrahedron, 2005, 61, 3289–3303.

115. Mills, R. J.; Taylor, N. J.; Sieckus, V. J. Org. Chem., 1989, 54, 4372.

116. Kessar, S. V.; Singh, P.; Vohra, R.; Kaur, N. P.; Venugopra, D. J. Org.

Chem., 1992, 57, 6716.

117. Conners, R.; Tran, E.; Durst, T. Can. J. Chem., 1996, 74, 221.

118. Larock, C. L.; Varaprath, S. US Pat. Appl., 1987, US 4650881. 119. Zenner, J. M.; Larock, R.C. J. Org. Chem., 1999, 64, 7312.

120. Warnell, J. H.; Shriner, R. L. J. Am. Chem. Soc., 1957, 79, 3165.

121. Suzuki,T.; Yamada, T.; Watanabe, K.; Katoh, T.;Biorg. Med.Chem.

Lett. 2005, 15, 2583-2585.

122. Ueura, K., Satoh, T., Miura, M., J. Org. Chem. 2007, 72, 5362.

123. Roy, H. N.; Sarkar, M. S. Synthetic Communications, 2005, 35, 2177.

124. Colonge, J.; Boisde, P. Bull. Soc. Chem. France, 1956, 1337.

125. Carter, R. H.; Colyer, R. M.; Hill, R. A.; Staunton, J. J. Chem. Soc. Perkin.

Trans. I., 1982, 1438.

126. Kendall, J. K.; Fisher, T. H. J. Org. Chem., 1989, 54, 4218.

127. Mal, D.; Bandyopadhyay, M.; Datta, K.; Murty, K. V. S. N. Tetrahedron,

1998, 54, 7525.

128. Mal, D.; Bandyopadhyay, M.; Sujit, K.; Datta, K. Tetrahedron Lett. 2000, 41,

1.

129. Kinder, M. A.; Kopf, J.; Margaretha, P. Tetrahedron, 2000, 56, 6763.

130. Bonadies, F.; DiFabio, R. J. Org. Chem., 1984, 49, 1647.

131. Loewenthal, H. J. E.; Pappo, R. J. Chem. Soc., 1992, 4799.

Page 327: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

140

132. Chatterjea, J. N.; Mukherjea, H. Experietia, 1960, 16, 439.

133. Chatterjea, J. N.; Mukherjee, H. J. Indian chem. Soc., 1960, 37, 379.

134. Chatterjea, J. N.; Mukherjee, H. J. Indian chem. Soc., 1960, 37, 443.

135. Yamato, M.; Hashigaki, K. Chem. Pharm. Bull., 1976, 24, 200.

136. Kabayashi, T.; Sci. Rept. Tohoku Univ., First Ser., 1942, 31, 73; C. A.,

1950, 44, 4013.

137. Haworth, R. D.; Pindred, H. K.; Jafferies, P. R. J. Chem. Soc., 1954, 3617.

138. Chatterjea, J. N. J. Indian chem. Soc., 1953, 30, 103.

139. Vorozhtsov, N. N.; Petushova, A. T. J. Gen. Chem. USSR, 1957, 27, 2282.

140. Tirdkar, R. B.; Usgoankar, R. N. Indian J. Chem., 1970, 8, 123.

141. Modi, R.; Usgoankar, R. N. Indian J. Chem., 1979, 17B, 360.

142. Rose, A.; Buu-Hoi, N. P.; Jacquinon, P. J. Chem. Soc., 1965, 6100.

143. Yoshikawa, H.; Taniguchi, E.; Maekawa, K. J. Pesticide Sci., 1980, 5, 1.

144. Sarkhel, B. K.; Srivasta, J. N. J. Indian chem. Soc., 1976, 53, 915; Ibid., 1977, 54, 925.

145. Tuanli Yao and Richard C. Larock, Tetrahedron Lett. 2002, 43, 7401.

146. Biagetti,M.; Bellina,F.; Carpita, A.; Stablie, P.; Rossi, R. Tetrahedron Lett.

2002, 58, 5023.

147. Yao, T.; Larock, R. C. J. Org. Chem. 2003, 68, 5936.

148. Abubakar, B. A.; Booth, L. B.; Edgar, A. E. J. Fluorine Chem., 1990, 47,

353.

149. Acke, D. R. J., Stevens, C. V., Green Chem., 2007, 9, 386.

150. Yamato, M.; Hashigaki, K.; Honda, E.; Sato, K.; Koyama, T. Chem. Pharm.

Bull., 1977, 25, 695.

151. Liu, D. Zhonghua Yixue Zashi, 1982, 62, 336.

152. Haworth, R. D.; Pindred, H. K.; Jafferies, P. R. J. Chem. Soc., 1954, 3617.

153. Nozawa, K.; Yamada, M.; Tsuda, Y.; Kawai, K. I.; Nakajima, S. Chem.

Pharm. Bull., 1981, 29, 2491.

154. Berti, G. J. Org. Chem., 1959, 24, 934.

155. Berti, G. Tethedron, 1958, 4, 393.

156. Muller, E. Chem. Ber., 1909, 42, 423.

Page 328: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

141

157. Ribbens, C.; Koninkl, N. V.; Fabrieken, P. v/h brocades-Stheeman

Pharmacia, 1960-61, 10, 9. C. A. 1962, 56, 7378.

158. Stadlbauer, W.; Ghobrial, N.; Kappe T. Z. Naturforsch, 1980, 35b, 892.

159. Prey, V.; Kerres, B.; Berbalk, H. Monatsh. Chem., 1960, 91, 774.

160. Berti, G.; Marsili, A.; Mini, V. Ann. Chim.(Rome), 1960,50, 669.

161. Birk, A. J.; Donovan, F. W. Australian J. Chem., 1953, 6, 360.

162. Colonge, J.; Boisde, P. Bull. Soc. Chim. France, 1956, 1337.

163. Maitte, P. Compt. Rend., 1954, 239, 1508.

164. Siegel, S.; Colburn, S. K.; Levering, D. R. J. Am. Chem. Soc., 1951, 73,

3163.

165. Shriner, R. L.; Knox, W. R. J. Org. Chem., 1951, 16, 1064.

166. Alder, E.; Magnusson, R.; Berggren, B. Acta Chem, Scand., 1960, 14, 539.

167. Grimshaw, J.; Haworth, R. D.; Pindred, H. K. J. Chem. Soc., 1955, 833.

168. Yamamoto, I. Agri. Biol. Chem. (Tokyo), 1961, 25, 400; C. A., 1961, 55,

670.

169. Thomas, O.L.; Jens, B. J.Nat. Prod., 1999, 62, 1182.

170. Stadler, M.; Anke, H.; Sterner, O. J. Antibiot., 1995, 48, 261.

171. Yoshikawa M.; Uchida E.; Chatani N.; Murakami N.; Yamahara J. Chem.

Pharm. Bull., 1992, 40, 3121.

172. Yoshikawa M.; Uchida E.; Chatani N.; Kobayashi H.; Naitoh Y.; Okuno Y.;

Matsuda H.; Yamahara J.; Murakami N. Chem. Pharm. Bull., 1992, 40,

3352.

173. Yoshikawa M.; Harada E.; Naitoh Y.; Inoue K.; Matsuda H.; Shimoda H.;

Yamahara J.; Murakami N.; Chem. Pharm. Bull., 1994, 42, 2225.

174. Yoshikawa M.; Matsuda H.; Shimoda H.; Shimada H.; Harada E.; Naitoh

Y.; Miki A.; Yamahara J.; Murakami N.; Chem. Pharm. Bull., 1996, 44,

1440.

175. Matsuda H.; Shimoda H.; Yoshikawa M. Bioorg. Med. Chem., 1999, 7,

1445.

176. Yamahara J.; Matsuda H.; Shimoda H.; Wariishi N.; Yagi N.; Mura- kami N.;

Yoshikawa M.; Folia Pharmacol. Jpn., 1995, 105, 365.

Page 329: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

142

177. Matsuda H.; Shimoda H.; Yamahara J.; Yoshikawa M. Biol. Pharm. Bull.,

1999, 22, 870.

178. Wang, Q.; Matsuda, H.; Matsuhira, K.; Nakamura, S.; Yuan, D.; Yoshikawa,

M. Biol. Pharm. Bull. 2007, 30(2), 388.

179. Rossi, R.; Carpita, A.; Bellina, F.; Stabile, p.; Mannina, L. Tetrahedron,

2003, 59, 2067.

180. Takeuchi, N.; Murase, M.; Ochi, K.;Tobinaga, S. J. Chem. Soc. Chem.

Comm., 1980, 13, 593.

181. Crombie, L.; Manzoor-i-Khuda, M.; Smith, R. J. D. J. Chem. Soc.,

1957,479.

182. Brain S. furniss “Vogel’s Text book of Practical Organic Chemistry”

Published by Longman Scientific & Technical England, 5th Ed., 1989, 410.

183. Brain S. furniss “Vogel’s Text book of Practical Organic Chemistry”

Published by Longman Scientific & Technical England, 5th Ed., 1989, 464.

184. Brain S. furniss “Vogel’s Text book of Practical Organic Chemistry”

Published by Longman Scientific & Technical England, 5th Ed., 1989, 1039.

185. Brain S. furniss “Vogel’s Text book of Practical Organic Chemistry”

Published by Longman Scientific & Technical England, 5th Ed., 1989, 1016.

186. C. Cope and Ronald Dean Smith, J. Org. Synthesis, 77, 4598 (1955).

187. Chatterjea, J. N.; Sengupta, S. C. Indian J. Chem., 1975, 13, 859.

188. Nozawa, K.; Nakajima, S.; Kawai, K. L. chem. Pharm. Bull., 1980, 28(4),

1112.

189. Thomas, O. L.; Jens, B. J. Nat. Prod. 1992, 62, 1182.

190. Odake, S.; Kam, C. M.; Narasimhan, L.; Poe, M.; Blake, J. T.; Krahenbuhl,

O.; Tschopp, J.; Powers, J. C. Biochemistry, 1991, 30, 2217.

191. Hudig, D.; Allison, N. J.; Kam, C. M.; Powers, J. C. Mol. Immunol., 1989,

26, 793. C. A., 111: 192909h.

192. Fratev, F.; Enchev, V.; Nikolov, P.; Polansky, O. E. Z. Naturforsch., A:

Phys. Chem., Komophys, 1984, 39A (11), 1143.

Page 330: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

143

193. Clark, M. T.; Gilmore, I. J. U. K. Pat. Appl.,GB 2207425. C. A, 111, 19479v.

194. Kerrigan, J. E.; Oleksyszyn, J.; Kam, C. M.; Selzler, J.; Powers, J. C. J.

Med. Chem., 1995, 38, 544.

195. Kam, C. M.; Kerrigan, J. E.; Plaskon, R. R.; Duffy, E. J.; Lollar, P.;

Suddath, F. L.; Powers, J. C. J. Med. Chem., 1994, 37, 1298.

196. Rama, N. H.; Saeed, A.; Hussain, M.T. J. Chem.Soc .Pak 1997, 19(4),

330.

197. Hussain, M.T.; Rama, N. H. Indian. J. Heterocyclic.Chem, 1998, 8, 99.

198. Khan, G. S., Rama, N. H., Noor, A., Kempe, R. & Qadeer, G. (2006). Z.

Kristallogr. New Cryst. Struct. 2006, 221, 153.

199. Garduño-Ramírez, M.L.; Delgado, G. Rev. Soc. Quím. Méx. 2003, 47, 160.

200. Waffo, T. P.; Fauconneau, B.; Deffieux, G., Huguet, F.; Vercauteren, J.;

Merillon, J. M. J. Nat. Prod. 1998, 61, 655.

201. Cotelle, N.; Vernier, J. L.; Catteau, J. P.; Pommery, J.; Wallet, J. C.;

Gaydou, E. M. Free Radical Biology & Medicine, 1996, 20, 35.

202. Molyneux, P. Songklanakarin J. Sci. Technol. 2004, 26, 211.

203. Baumann, J.; Wurn, G.; Bruchlausen, F. V. Naunyn-Schmiedebergs Arch.

Pharmacol. 1979, 308, R27.

204. Hiller, P. L.; Hodd, K. O.; Wilson, R. L. Chem. Biol. Interact. 1983, 47, 293.

205. Williams, B.; Cuvelier, M.E.; Berset, C. Lebensmittel Wissenschaft Technol.

1995, 28, 25.

206. Böger, P.; Wakabayashi, K.; Hirai, K.; Herbicide Classes in Development,

Springer-Verlag: Berlin, Germany, 2002, p. 364 (ISBN 0540431470).

207. Heap, I.; http://www.weedscience.org, accessed Sept 21, 2005.

208. Beck-Sague, C. M.; Jarvis, W. R.; J. Infect. Dis. 1993, 167, 1247.

209. Diekema, D. J.; Messer, S. A.; Brueggemann, A. B.; Coffman, S. L.; Doern,

G. V.; Herwaldt, L. A.; Pfaller, M. A.; J. Clin. Microbiol. 2002, 40, 1298.

210. Fridkin, S. K.; Jarvis, W. R.; Clin. Microbiol. Rev. 1996, 9, 499.

211. Wenzel, R. P.; Clin. Infect. Dis. 1995, 20, 1531.

212. Wingard, J. R.; Clin. Infect. Dis. 1995, 20, 115.

Page 331: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

144

213. Irvine, N. M.; Ricks, M. J.; Ross, R.; Bryan, K.; Klittich, C. J. R.; CAN 142:

392441; Dow Agro sciences LLC: PCT Int. Appl., 2005033095, 2005.

214. Yamaguchi, I.; Fujimura, M.; J. Pestic. Sci. 2005, 30, 67.

215. Zhi-Jin, F.; Bin, L.; Xiu-Feng, L.; Bin, Z.; Chang-Ling, L.; Zheng-Ming, L.;

Chemical Journal of Chinese Universities, 2004, 25, 663.

216. Waitz, J.A; Deuve, C.G. Annu. Rep. Med. Chem., 1972, 7, 109.

217. De Clercq, E.; M. Luczak, J. C.; Reepmeyer, K.; Kirk, L.; Cohen, L. A. Life

Sci. 1975, 17, 187.

218. De Clercq, E.; Holy, A.; Rosenberg, I.; Sakuma, T.; Balzarini, J.; Maudgal,

P. C. Nature, 1986, 323, 464.

219. De Clercq, E.; Yamamoto, N.; Pauwels, R.; Baba, M.; Schols, D.;

Nakashima, H.; Balzarini, J.; Debyser, Z.; Murrer, B. A.; Schwartz, D.;

Thornton, D.; Bridger, G.; Fricker, S.; Henson, G.; Abrams, M.; Picker, D.

Proc. Natl. Acad. Sci. U.S.A. 1992, 89, 5286.

220. Balzarini, J.; Naesens, L.; Slachmuylders, J.; Niphuis, H.; Rosenberg, I.;

Schellekens, H.; De Clercq, E. AIDS, 1991, 5, 21.

221. Bridger, G. J.; Skerlj, R. T.; Padmanabhan, S.; Martellucci, S. A.; Henson,

G. W.; Abrams, M.J.; Joao, H.C.; Witvrouw, M.; De Vreese, K.; Pauwels,

R.; Erik De Clercq, E. J. Med. Chem. 1996, 39, 109.

222. De Clercq, E.; Yamamoto, N.; Pauwels, R.; Balzarini, J.; Witvrouw, M.; De

Vreese, K.; Debyser, Z.; Rosenwirth, B.; Peichl, P.; Datema, R.; Thornton,

D.; Skerlj, R.; Gaul, F.; Padmanabhan, S.; Bridger, G.; Henson, G.; Abrams,

M. Antimicrob. Agents Chemother. 1994, 38, 668.

223. Bridger, G. J.; Skerlj, R. T.; Thornton, D.; Padmanabhan, S.; Martellucci, S.

A.; Henson, G. W.; Abrams, M. J.; Yamamoto, N.; De Vreese, K.; Pauwels,

R.; De Clercq, E. J. Med. Chem. 1995, 38, 366.

224. Joao, H. C.; De Vreese, K.; Pauwels, R.; De Clercq, E.; Henson, G. W.;

Bridger, G. J. J. Med. Chem. 1995, 38, 3865.

225. De Vreese, K.; Reymen, D.; Griffin, P.; Steinkasserer, A.; Werner, G.;

Bridger, G. J.; Este, J.; James, W.; Henson, G. W.; Desmyter, J.; Anne, J.;

De Clercq, E. Antiviral Res. 1996, 29, 209.

Page 332: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

145

226. Janeba, Z.; Balzarini, J.; Andrei, G.; Snoeck, R.; De Clercq, E.; Robins, M.

J. J. Med. Chem. 2005, 48, 4690.

227. Ching, L. L.; Vlad, R.; Man-Fung, Y.; Thierry, P. Lancet, 2003, 362, 2089.

228. De clercq, E. J. Clin. Virol. 2004, 30, 115.

229. Ganem, D.; Prince, A. M. N. Engl. J. Med. 2004, 350, 1118.

230. Humphries, J. C.; Dixon, J. S. Intervirology 2003, 46, 413.

231. Bracke, M. E.; Van Larebeke, N. A.; Vyncke, B. M.; Mareel, M. M. Br J.

Cancer. 1991, 63, 867.

232. Romijin, J. C.; Verkoelen, C. F.; Schroeder, F. H. Prostate, 1988, 12, 99.

233. Bracke, M. E.; Boterberg, T.; Bruynell, E. A.; Mareel, M. M. (Eds. Brooks, S.

and Schumzcher, U.) Hannover press, Totowa, NJ, 1999.

234. Steelant, W. F.; Goeman, J. L.; Philippe, J.; Oomem, L. C.; Hilkens, J.;

Krzewinski-Recchi, M. A.; Huet, G.; Van der Eycken, J.; Delannoy, P.;

Bruynell, E. A.; Mareel, M. M. Int. J. Cancer, 2001, 92, 527.

Page 333: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

316

REFERENCES (PART TWO)

1. Bamberger, E.; Gruyter, P. de., Ber. 1893, 26, 2385.

2. Pellizzari, G., Gazz. Chim. Ital., 1911, 41(20), 93.

3. Potts, K.T., Chemical Review, 1961, 61, 87.

4. Kubata, S.; Uda, M., Chem. Pharm. Bull., 1972, 20, 2096.

5. Kubata, S.; Uda, M., Chem. Pharm. Bull., 1973, 21, 1342.

6. Hoggarth, E. J., Chem. Soc., 1949, 1163.

7. Maquestiau, A.; Haverbeke, Y.V.; Flammang, R.; Mispreuve, H.,

Org. Mass Spectrum, 1977, 12, 2005.

8. Elguro, J.; Marzine, C.; Katritzky, A.R.; Linda, P., “The

Tautomerism of Heterocycles”, Academic Press, Inc., London,

1976.

9. Staab, H.A., Chem. Ber., 1956, 89, 1927.

10. Atkinson, M.R.; Parkes, E.S.; Polya, J.B., J. Chem. Soc., 1954, 4256.

11. Potts, K.T., J. Chem. Soc., 1954, 1361.

12. Pavia, D.L.; Lampman, G.M.; Kriz, G.S., Introduction to

Spectroscopy, Brooks/Cole, Thomson Learning, 2001.

13. Rollas, S.; Buyuktimkin, S.; Cevikbas, A., Arch. Pharm., 1991, 324, 189.

14. Moshen, A.; Omar, M.E.; Osman, S.A., Pharmazie, 1973, 28, 30.

15. Ulusoy, N.; Ates, O.; Kucukbasmaci, O.; Kiraz, M.; Yegenoglu, Y.,

Monatshefte fur Chemie, 2003, 134, 465.

16. Singh, G.; Felix, S. P., J. Hazardous Materials, 2002, A90, 1.

17. Zamani, K.; Faghihi, K.; Sangi, M.R.; Zolgharnein, J., Turk. J.

Chem., 2003, 27, 119.

18. Bala. S.; Gupta, R.P.; Sachdeva, M.L.; Singh, A.; Pujari, H.K.,

Indian J. Chem., 1978, 16B, 481.

19. Mohan, J., Indian J. Chem., 1983, 22B, 270.

20. El-masry, A.H; Fahmy H.H.; Ali Abdelwahed, S.H., Molecules, 2000, 5, 1429.

Page 334: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

317

21. Orabi, A.S.; Moneim, M.A.; El-Din salem E.; El-Din Abdel-Fattah, M.,

Polish J. Chem., 2000, 74, 1675.

22. Martin, G.; German Patent, 2,240,043. (Cl. C 07 d) March(1973);

Chem. Abstr., 1973, 78, 136302t.

23. Holla, B. S.; Veerendra, B.; Shivananda, M. K.; Poojary, B. Eur. J.

Med. Chem. 2003, 38, 759.

24. Duran, A.; Dogan, H. N.; Rollas S., Farmaco 2002, 57, 559.

25. Parmar, S.S.; Rastogi, V.K.; Agarwal, V.K.; Sinha, J. N.;

Chaudhari, A., Can. J. Pharm. Soc., 1974, 9, 107.

26. George, T.; Mehta, D.V.; Tahilramani, R.; Davvid, J.; Talwalker,

P.K., J. Med. Chem., 1971, 14, 335.

27. Yale, H.L.; Piala, J.J., J. Med. Chem., 1966, 9, 42.

28. Shah, M.H.; Mhasalkar, M.Y.; Patki, M.V.; Deliwala C.V.; Sheth,

U.K., J. Pharm. Sci., 1969, 58, 1398.

29. Kuranari, M.; Takeuchi, H., Chem. Abstr., 1967, 21918d, 67.

30. Burch, H.A.; Smith, W.O., J. Med. Chem., 1966, 9, 405.

31. Abdou, N.A.; Amin, F.M.; Mansoura, J. Pharm. Sci., 1990, 6, 25.

32. Mhasalkar, M.Y.; Shah, M.H.; Nikam, S.T.; Anantanarayanan,

K.G.; Deliwala, C.V., J. Med. Chem., 1970, 13, 672.

33. Mohasalkar, M.Y.; Shah, M.H.; Nikam, S.T.; Anantanarayanan,

K.G.; Deliwala, C.V., J. Med. Chem., 1971, 14, 260.

34. Kuranari, M.; Takeuchi, H., Chem. Abstr., 1967, 21918d, 67.

35. Mir, I.; Siddiqui M.T.; Comrie, A., Tetrahedron, 1970, 26, 5235.

36. Mishra, R.K.; Tewari, R.K., et al, J. Indian Chem. Soc., 1991, 68,

110.

37. Suman, S.P.; Bahel, S.C., J. Indian Chem. Soc., 1979, 56, 374.

38. Buscemi, S.; Vivona, N.; Caronna, T., J. Org. Chem., 1996, 61,

8379.

39. Garoufalias, S. P., Pouli, N., Marakos, P., Chytyroglou-Ladas, A.

Farmaco., 2002, 57, 973

40. Yu, B.R. Yoo. M.Y. Suk. Y-Man.; Hong, S-Gyn.; Jung, I.N., Bull.

Korean Chem. Soc., 1998, 19(3), 358.

41. Paulvannan, K.; chen, T.; Hale, R., Tetrahedran, 2000, 56, 8071.

42. Catarzi, D.; Colotta, V.; et. al., J. Med. Chem.., 2000, 43, 3824.

Page 335: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

318

43. Zamani, K. ; Faghihi, K. ; Sangi, M. R. ; Zolgharnein, J. Turk J

Chem., 2003, 27, 119.

44. Saksena, R.K.; Rana, Y. Ind. J. Chem., 1987, 26B, 290.

45. Kruse, L. I.; Finkelstein, J. A. (Smith Kline Bechmenn corp.). Eur.Pat.

Appl., 1990, EP-359505 (Cl. CO7D401/06), CA/113:115311w.

46. Selander, E. H.; et al, Acta Pharm. Sci. , 1987, 24 (3), 123.

47. Anon(USA). Res. Discl. , 278, 365, Chem. Abstr. 1987, 108,

131697d.

48. Amir, M.; Shahani.; Shalini. Indian J. Heterocycl.Chem., 1998, 8(2), 107.

49. El-Din, N. S.; El-Fatatry, H. M.; El-Hamamsy, M., Alexandria J.

Pharm. Sci., 1998, 12, 15.

50. Singh, C.A.; Langer, T.C.; Sharma, S.K., J. Indian. Chem. Soc.,

1989, 66, 122.

51. Vamvakides, A.; Pharm. Fr., 1990, 48, 154.

52. Grambaryan, G.S. Izv. S. Kh. Nauk., 1983, 26, 40.

53. Misato, T.; Ko, K.; Honma, Y.; Konno, K.; Taniyama, E. Japan

Patent, 77-25028 (A01N 9/12); Chem. Abstr., 1977, 87, 147054.

54. Martin, R.J.; Tu, L.N.; Muthuvelu, T. Eur. Patent, EP 337815;

Chem. Abstr., 1990, 112, 198386c.

55. Demirbas, N.; Demirbas, A. ; Sancka, K., Turk. J. Chem., 2002, 26,

801.

56. Heeres, J.; Hendrickx, R.; Van Custem, J. J. Med. Chem., 1983, 26,

611.

57. Katritzky, A.R.; Pastor, A.; Voronkov, M.; Steel, P.J., Org. Lett.,

2000, 2, 429.

58. Katritzaky, A.R; Feng, M.Qi, D.; Zhang, G.; Griffith, M.C.; Watson,

K.; Org. Lett., 1999, 1,1189.

59. Heeres, J.; Backx, L.J.J.; Van Custem, J., J. Med. Chem., 1984, 27,

894.

60. Shafie, A.; Nassian, F.; Reghavi, N., J. Heterocycl. Chem., 1992, 29,

1863.

61. Rkovskay, L. G.; Knysh, F. G.; Rogulchemko, G. L ., et al, Farm. 2h,

(Kiev), 1989, 5 , 67. Chem l. Abstr., 1990, 112(1), 98233 a.

Page 336: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

319

62. Patil, S. A.; Bodiger, B. M.; Kudasi, S. M.; Klkerni, V.H., J. Indian

Chem. Soc., 1984, 61 (8), 713.

63. Srivastava, S. K.; Pathak, R. B.; Bahel, S. C., J. Indian Chem.

Soc., 1989, 66 (3), 210.

64. Kawanishi, Wiroyuki, Morimoto, Hiroshi, Nakano, Takao,

Watanabe, Tatasya, Oda, Kuniyuki, Tsujhara, Heterocycles, 1998, 49,

181.

65. Wierzbicki, P. Hugon, J. C. Poingnant, (ADIR) Eur .Pat. Appl. EP. 199,

641(Cl. CO7 D 487/04), (1986).

66. Hasegawa, K.; Kariyama, T., et al, (Onta Pharmaceutical Co.Ltd.)

Jpn. Kokai Tokyo JP 61, 148, 176 [86, 148, 176]. , (Cl. CO7 D 401/12),

1986.

67. Curtius, T., et al., Chem. Ber., 1998, 41, 3168.

68. Ebied, M. Y.; Ashmawi, M. I. AL.; Abbas, E.S., et al, Egypt. J.

Pharm. Sci., 30 (1-40), 339, Chem. Abstr., 1989, 112, 235265 K.

69. Lewwnstein, M.J., U. S. 2, 683,106, Chem. Abstr., 1954, 48, 13175 b.

70. Somerai, T.; Szilagyi, L.; Horvath, S., Arch. Pharm., 139 (3), 238.

Chem. Abstr. 1986, 105: 6416j.

71. Kane, M. J.; Miller, F. P., Eur. Pat. 276, 793 (Cl. A 61 K 31/44),

Chem. Abstr., 1988, 109, 1704431.

72. Velaqvez, S.; Alvarez, R.; Pervez, C., Antiviral chem. Chemother.,

1998, 9 (6), 481.

73. Latge, J. P. Aspergillus fumigatus and aspergillosis., Clin.

Microbiol.ReV., 1999, 12, 310.

74. Steenbergen, J. N.; Casadevall, A., J. Clin. Microbiol, 2000, 38,

1974.

75. Arikan, S.; Rex, J.H. Nystatin LF (Aronex/Abbott). Curr. Opin.

Invest. Drugs, 2001, 2, 488.

76. Denning, D. W., Echinocandins: a new class of antifungal. J.

Antimicrob. Chemother. 2002, 49, 889.

77. Birnbaum, J. E., Pharmacology of the allylamines. J. Am. Acad.

Dermatol. 1990, 23, 782.

78. Hoffman, H. L.; Ernst, E. J.; Klepser, M. E., Expert Opin. InVest.Drugs,

2000, 9, 593.

Page 337: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

320

79. Casalinuovo, I. A.; Di Francesco, P.; Garaci, E., Eur. ReV. Med.

Pharmacol. Sci. 2004, 8, 69.

80. Chandrasekar, P. H.; Manavathu, E., Voriconazole: A second

generation triazole. Drugs Today (Barc.) 2001, 37, 135.

81. Herbrecht, R., Int. J. Clin. Pract. 2004, 58, 612.

82. Arikan, S.; Rex, J.H., Curr. Opin. Invest. Drugs, 2002, 3, 555.

83. Schmidt, R. R.; Heermann, D., Chem. Ber., 1981, 114, 2825.

84. Hanna, N. B.; Dimitrijevich, S. D.; Larson, S. B.; Robins, R. K.;

Revankar, G. R., J. Heterocycl. Chem. 1988, 25, 1857.

85. Witkowski, J. T.; Robins, R. K.; Sidwell, R. W.; Simon, L. N., J.

Med. Chem., 1972, 15, 1150.

86. Camarasa, M.-J.; De Las Heras, F. G., J. Heterocycl. Chem.,

1983, 20, 1307.

87. Allison, M.C.; Howatson, A.G.; Torrance, C.J.; Lee, F.D.; Russel,

R.I.N., Engl. J. Med., 1992, 327, 749.

88. Adachi, Chihaya, Balao, March-A, Thompson, Mark-E, J. appl.

Phys. 2001, 90(10), 5048.

89. Choi, U.; Kim, T.; Jung, S.; Kim, C., Bullention of the Korean Chemical

Society, 1998, 19(3).

90. Izatt, Lind, Bruening,, Analytical Chemistry. 1998, 60, 1694.

91. Akira, K.; Jakeshi, H., (Sakai chem. Industry Co.Ltd.) Japan’s

Kokai, 7362, 634(Cl. 12 A 82), 01, Sept., 1973.

92. Stephanyan, E.S.; Delo, V., Chem. Abstr., 1966, 64, 4146.

93. Kotone, A.; Hori, T., Chem. Abstr., 1974, 80, 62968g.

94. Schofield, K.; Grimmett, M.R.; Keene, B.R.T., Heteroaromatic

Nitrogen compounds. The Azoles Cambridge University Press,

1976, 34.

95. Lixue, Z.; et al, Gaodeng Xuexiao Huaxue Xuebao, 1990, 11,

148.

96. Gambaryan, G.S., Izv. S. Kh. Nauk., 1983, 26, 40.

97. Grimmel, H.W.; Morgan, J.F., Chem. Abstr., 1951, 45, 2683c.

98. Allen, W.W., U.S. Patent, 1954, 2670282. Linser, H.; Kiermayer, O.

Planta, 1957, 49, 498.

99. Ainsworth, C., Jones, R.G., J. Am. Chem. Soc., 1955, 77, 621.

Page 338: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

321

100. Grundmann, C. J.; Kreutzberger, A., J. Am. Chem. Soc., 1957, 79,

2839.

101. Grundmann, C. J.; Ratz, R. F. W., J. Org. Chem., 1956, 21, 1037.

102. Grundmann, C. J.; Ratz, R. F. W., U.S. patent, 2,800,486 (July 23,

1957); Chem. Abstr., 1957, 51, 18009a.

103. Widman, O., Ber. 1893, 26, 2612.

104. Pellizari, G.; Soldi, A., Gazz. Chim. Ital., 1905, 35, 373.

105. Wohl., A.; Marckwald, W., Ber., 1889, 22, 576.

106. Hoggarth, E., J. Chem. Soc., 1949, 1163.

107. Maertens, J.A. Clin. Microb. Infect. 2004, 10, 1.

108. Jones, R. G.; Ainsworth, C., J. Am. Chem. Soc., 1955, 77, 1538.

109. Ragenovic, C. K., Dimova, V., Kakurinov, V., Molnar, D. G.,

Buzarovska, A. Molecules, 2001, 6, 815.

110. Mustafa, S. M., Nair, V. A., Chittoor, J. P., Krishnapillai, S. Mini-

Reviews in Organic Chemistry, 2004, 1, 375.

111. Young, G.; Witham, E., J. Chem. Soc., 1900, 77, 224.

112. Potts, K. T.; Huseby, R. M., J. Org. Chem., 1966, 31, 3528.

113. Vainilavicins, P.; Smicius, R.; Jakubkiene, V.; Tunkevicius, S.,

Monatsh. Chem., 2001, 132, 825.

114. Reid, J. R.; Heindel, N. D., J. Heterocycl. Chem., 1976, 13, 925.

115. Al-Soud, Y. A.; Al-Masoudi, N.A., J. Braz. Chem. Soc., 2003, 14, 790.

116. Farghaly, A. A. H., J. Chin. Chem. Soc., 2004, 51, 147.

117. Schulze, K.; Richter, C.; Ludwig, R.; Klatt, K. Z., Chem., 1988,

28(8), 288.

118. Patil, S.A.; Badiger, B.M.; Kudari, S.M.; Kulkarni, V.H., J. Ind.

Chem. Soc., 1984, 61(8), 713.

119. Prasad, A.R.; Rao, A.N.; Ramalingam, T.; Sattur, P.B., Ind. Drugs,

1988, 25(7), 301.

120. Kroeger, C.F.; Tenor, E.; Beyer, H., Justus Liebigs Ann. Chem.,

1961, 643, 128.

121. Kroeger, C.F.; Beyer, H.; Busse, G., Justus Liebigs Ann. Chem., 1960,

637, 135.

122. Fangzhen, L.; Ruqi, H.; Zongni, W.; Xinyu, Y., Huaxue Tongbao, 1996, 10, 38.

Page 339: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

322

123. Fromm, P.; Kappeler, R.; Feniger, M.; Krauss, P.;Schwanenfeld, M.;

Wetternik, L., Ann., 1926, 447, 294.

124. Weidinger, H.; Kranz, J., Chem. Ber., 1963, 96, 1059.

125. Yeung, K.; Farkas, M.E.; Kadow, J.F.; Meanwell, N.A.,

Tetrahedron Lett., 2005, 46, 4329.

126. Contour-Galcera, M.; Sidhu, A.; Plas, P.; Roubert, P., Bioorg.

Med. Chem. Lett., 2005, 15, 3555.

127. Ahmad, R.; Zia-ul-Haq, M.; Jabeen, R.; Duddeck, H., Tr. J. Chem.,

1996, 20, 186.

128. Bentiss, F.; Lagrenee, M.; Barby, D. Tetrahedron Lett., 2000, 41,

1539.

129. Katritzky, A. R.; Qi, M. ; Feng, D. ; Zhang, G. ; Griffith, M. C. ;

Watson, K. Org., Lett., 1999, 1, 1189.

130. Boeglin, D. ; Cantel, S. ; Heitz, A. ; Martinez, J. ; Fehrentz, J.-A.

Org. Lett., 2003, 5, 4465.

131. Samanta S. K.; Yli-Kauhaluoma, J. J. Comb. Chem., 2005, 7,

142.

132. Kingston, H. M.; Haswell, S. J.; Microwave-Enhanced Chemistry

Fundamentals, Sample Preparation and Application, American

Chemical Society: Washington, D. C., 1997.

133. Loupy, A. Microwaves in Organic Synthesis, Wiley-VCH:

Weinheim, 2002.

134. Heyes, B. L. Microwaves Synthesis; Chemistry at the Speed of

Light, CEM Publ: Matthews, NC, 2002.

135. Perreaux, L.; Loupy, A. Tetrahedron, 2001, 57, 9199.

136. Lidström, P.; Tierney, J. ; Wathey, B. ; Westman, J. Tetrahedron,

2001, 57, 9225.

137. Fini, A.; Breccia, A. Pure Appl. Chem., 1999, 71, 573.

138. Abramovich, R. A. Org. Prep. Proced. Int., 1991, 23, 685.

139. Caddick, S. Tetrahedron, 1995, 51, 10403.

140. Strauss C. R.; Trainor, R. W. Aust. J. Chem., 1995, 48, 1665.

141. Bose, A. K. ; Banik, B. K. ; Lavlinskaia, N.; Jayaraman, M. ; Manhas,

M. S. Chemtech., 1997, 27, 18.

142. Strauss, C. R. Aust. J. Chem., 1999, 52, 83.

Page 340: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

323

143. Kidwai, M.; Kumar, R. Gazz. Chim. Ital., 1997, 127, 203.

144. Larhed, M.; Moberg, C.; Hallberg, A. Acc. Chem. Res., 2002, 35, 717.

145. Nüchter, M. ; Müller, U. ; Ondruschka, B. ; Lautenschläger, W.

Chem. Eng. Technol., 2003, 26, 1208.

146. Nüchter, M. ; Ondruschka, B. ; Bonrath, W. ; Gum, A. Green Chem.,

2004, 6, 128.

147. Kappe, C. O.; Stadler, A. Microwave-Assisted Combinatorial

Chemistry, in Microwaves in Organic Synthesis, A. Loupy, (ed.),

Wiley-VCH, 2002, p. 405.

148. Kappe, C. O. Curr. Opin. Chem. Biol., 2002, 6, 314.

149. Kappe, C. O. Am. Lab., 2001, 33, 13.

150. Molteni, V.; Ellis, D. A. Curr. Org. Synth., 2005, 2, 333.

151. Katritzky, A. R.; Pastor, A.; Voronkov, M.; Steel, P. Org. Lett.,

2000, 2, 429.

152. Koshima, H.; Hamada, M.; Tani, M. ; Iwasaki, S. ; Sato, F.

Heterocycles, 2002, 57, 2145.

153. Rostamizadeh, S. ; Tajik, H. ; Yazdanfarahi, S. Synth. Commun.,

2003, 33, 113.

154. Li, D.; Bao, H.; You, T. Heterocycles, 2005, 65, 1957.

155. Siddiqui, A. ; Arora, A. ; Siddiqui, N. ; Misra, A. Indian J. Chem.,

2005, 44B, 838.

156. Kidwai, M.; Mohan, R. J. Korean Chem. Soc., 2004, 48, 177.

157. Kornis, G., 1,3,4-Thiadiazoles. Comprehensive Heterocyclic

Chemistry, Katritzky, A. R.; Rees, C. W., Eds., Pergamon Press,

Oxford, 1984, 6(4B), 545.

158. Shouji, E.; Buttry, D. A., J. Phys. Chem. B, 1998, 102, 1444.

159. Katritzky, A. R.; Borowiecka, J.; Fan, W.-Q.; Brannigan, L. H., J.

Heterocycl. Chem., 1991, 28, 1139; Katritzky, A. R.; Wang, Z.;

erman, R. J. O., J. Heterocycl. Chem., 1990, 27, 139.

160. Bats, J. W., Acta Crystallogr., Sect. B: Struct. Crystallogr. Cryst.

Chem., 1976, 32, 2866.

161. U.S.3, 90,537(1975), Lubrizol Corp., inv.: D. E. Riple; Chem.

Abstr. 1976, 84, 7432k.

Page 341: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

324

162. Oleson, J.H.; Tory, W.P.; Williams, J.H., J. Am. Chem. Soc., 1995, 77,

6713.

163. Hazaa, B.A.A.; Ashour, F.; Shafik, R.M., Pharmazia, 1980, 35,

324.

164. Knysh, F.G.; Mazur, I.A.; Stets, V.R.; Sevenkora, N.N., Farm. Zh.

(Kier), 1986, 2, 49.

165. Peter, S., J. Indian. Chem. Soc., 1991, 68, 245.

166. Halsted, R.A.; Jessup, P., U.S. 4, 623, 474 (C1, 252-475: Clam

133/38) (1986); Chem. Abstr., 1988, 107, 170619d.

167. Dogan, H.N.; Rollas S.; Erdeniz, H., Il Farmaco, 1998, 53, 462.

168. Mamolo, M.G.; Vio, L.; Banfi, E., Il Farmaco, 1996, 51, 71.

169. Dogan, H.N.; Duran, A.; Rollas, S.; Sener, G.; Uysal, M.K.; Gülen, D.,

Bioorg. Med. Chem., 2002, I0, 2893.

170. Palaska, E.; Sahin, G.; Kelicen, P.; Durlu, N.T.; Altinok, G., Il

Farmaco, 2002, 57, 101.

171. Foroumadi, A.; Kiani, Z.; Soltani, F., Il Farmaco, 2003, 58, 1073.

172. Chufán, E.E.; Pedregosa, J.C.; Baldini, O.N.; Bruno-Blanch, L., Il

Farmaco, 1999, 54, 838.

173. Krutovskikh, G.N.; Rusanov, A.M.; Gornaeva, G.F.; Vartanyan,

L.P.; Kolesova, M.B., Kimiko-Farmatsevticheskii Z., 1977, 11, 48.

174. Chou, J.-Y.; Lai, S.-Y.; Pan, S.-L.; Jow, G.-M.; Chern, J.-W.; Guh, J.-

H., Biochem. Pharmacol., 2003, 66, 115.

175. Oleson, J.J.; Sloboda, A.; Troy, W.P.; Halliday, S.L.; Landes,

M.J.; Angier, R.B.; Semb, J.; Cyr. K.; Williams, J.H., J. Am. Chem.

Soc., 1955, 77, 6713.

176. Davis, M., Org. Compd. Sulphur, Selenium, Tellurium, 1979, 5,

440.

177. Lu, K.; Loo, T., Cancer. Chemother. Pharmacol., 1980, 4, 275.

178. Kusterer, K.; Szabo, S., Eur. J. Pharmacol., 1987, 141, 7.

179. Roblin, R.O.; Clapp, J. W., J. Am. Chem. Soc., 1950, 72, 4890.

180. Maren, T.H., Physiol.Rev., 1967, 47, 595.

181. Maren, T.H., Biochemistry and Genetics to Physiology and Clinical

Medicine, F. Botr6 et al. Eds., VCH, Weinheim, 1991, 186.

Page 342: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

325

182. Maren, T.H.; Clare, B.W.; Supuran, C.T., Roum. Chem. Quart.

Rev., 1994, 2, 259.

183. Maren, T.H., Glaucoma, J., 1995, 4, 49.

184. Supuran, C.T.; Lepadatu, C.I.; Olar, R.; Meghea, A.; Brezeanu, M.,

Rev.Roum.Chim., 1993, 38, 1509.

185. Alzuet, G.; Casanova, J.; Ramirez, J.A.; Borrhs, J.; Carugo, J.

Inorg.Biochem., 1995, 57, 219.

186. Thimmaiah, K.N.; Chandrappa, G.T.; Lloyd, W.D.; Parkanyi, C.,

Inorg. Chim. Acta, 1985, 107, 1.

187. Kuodis, Z., A. Rutavichyus, and S. Valiulene, Salts of 2,5-

dimercapto-1,3,4-thiadiazole. Chem. Heterocycl. Compd., 2000,

36(5), 598.

188. Gao, S. and Y. Yin., Huaxue Tongbao, 1998, 3, 48.

189. Graf, H.-J.; Schaefer, V.; Schilling, K.; Schudok, C., Vulcanizing

agents. German Patent No. 4424680. Germany. Rhein-Chemie

Rheinau G.m.b.H. January 18, 1996. CA 124:204691.

190. Crouch, R.L., World Patent No. 9858039. U.S. Chemonics Fire-

Trol, Inc. December 23, 1998.

191. Crouch, R.L.; Burchert, D. D. European Patent No. 693304. U.S.

Chemonics Industries, Inc. January 24, 1996.

192. Kegeler, G.H., and H.L. Vandersall. U.S. Patent No. 4606831.

U.S. Monsanto Co. June 19, 1986.

193. Fujiki, K., Japanese Patent No. 09328573. Japan. Bridgestone

Corporation. December 22, 1997.

194. Jones, E., Warren, N.E. U.S. Patent No. 5216053. U.S. Dow Chem.

Co. June 1, 1993.

195. Ahmed, M.J.; Al-Mosaddeque, M., Talanta, 2001, 55(1), 43.

196. Ahmed, M.J.; Israt, J.; Saera, B, Anal. Sci. Int. J. Jpn. Soc. Anal.

Chem., 2002, 18(7), 805.

197. Ben-Bassat, A.H.; Alony, T., Anal. Chim. Acta 1976, 83(1), 403.

198. Li, C.; James, B.D.; Rumble, J.; Magee, R.J., Mikrochim Acta

1988, 3(1-6), 175.

199. Maxwell, T.J.; Smyth, W.F., Electroanalysis, 1996, 8(8-9), 795.

Page 343: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

326

200. Buckl, H. 1984. German Patent No. 3231982. Germany. Sued-

Chemie A.-G. March 1, 1984.

201. Hudson, M.J.; Hassan, M.B.; Tiravanti, G., Stud. Environ. Sci. 29, 529.

202. Lessi, P.; Filho, N.L.D.; Moreira, J.C.; Campos, J.T.S., Anal. Chim.

Acta, 1996, 327(2), 183.

203. Domenico, P., U.S. Patent No. 6086921. U.S. Winthrop-University

Hospital. July 11, 2000. Abstract from TOXCENTER 2000:166908.

204. Hasegawa, Y.; Dohtain, M.; Ito, H.; Dohke, G., 1973. Japanese

Patent No. 7338141. Japan. Kumiai Chemical Industry Co., Ltd.

November 15, 1973. Abstract from TOXCENTER 1974:74605.

205. Charkit Chemical Corp. Undated. CAS No. 1072-71-5. March 26, 2002.

206. Inoue, E.; Kokado, H.; Yamaguchi, T.; Tokunaga, Y., German

Patent No. 2359843. Germany. Fuji Photo Film Co., Ltd. June 6,

1974.

207. Okutsu, E.; Yamada, K.; Hirano, S., Japanese Patent No.

97297379. Fuji Photo Film Co., Ltd. November 18, 1997. Abstract from

TOXCENTER 1997:213191.

208. Ohta, M., J. Pharm. Soc. Jp., 1952, 72, 1536.

209. Arnalt, F., Ber., 1922, 55, 343.

210. Ban, S., J. Pharm. Soc. Jp., 1954, 74, 695.

211. Whitehead, C.W.; Traverso, J.J., J. Am. Chem. Soc., 1955, 77,

5872.

212. Weiss, M. J.; Remers, W. A.; Gibs, G.J., J. Heterocycl. Chem. 1969, 6, 835.

213. Bernstein, J.; Yale, H. L.; Losee, K.; Holsing, M.; Martins, J.; Lott, W.

A. J. Am. Chem. Soc. 1951, 73, 906.

214. Ohta, M., J. Pharm. Soc. Jp., 1953, 73, 1127.

215. Barton; Ollis. omprehensive Organic Chemistry, 1979, 14, 405.

216. Rasmussen, P. B.; Pedersen, U.; Thomsen, I.; Yde, B.;

Lawesson, S.-O. Bull. Soc. Chim. Fr., 1985, 62.

217. Sumpter, W.C. Chem. Rev. 1954, 34, 407.

218. Popp, F.D. Adv. Heterocycl. Chem. 1975, 18, 1.

219. Shvekhgeimer, M.G.A. Chem. Heterocycl. Compd.(Engl.

Transl.).1996, 32, 249.

Page 344: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

327

220. Guo, Y.; Chen, F. Zhongcaoyao 1986, 17, 8. (CA104:213068f).

221. Yoshikawa, M.; Murakami, T.; Kishi, A.; Sakurama,T.; Matsuda, H.;

Nomura, M.; Matsuda, H.; Kubo,M. Chem. Pharm. Bull. 1998, 46, 886.

222. Bergman, J.; Lindström, J.O.; Tilstam, U. Tetrahedron.1985, 41,

2879.

223. Wei, L.; Wang, Q.; Liu, X. Yaowu Fenxi Zazhi 1982, 2, 288. (CA

98:95726b)

224. Ischia, M.; Palumbo, A.; Prota, G. Tetrahedron 1988, 44, 6441.

225. Palumbo, A.; Ischia, M.; Misuraca, G.; Prota, G. Biochim. Biophys. Acta

1989, 990, 297.

226. Halket, J.M.; Watkins, P.J.; Przyborowska, A.; Goodwin, B.L.;

Clow, A.; Glover, V.; Sandler, M. J. Chromatogr. 1991, 562, 279.

227. Kapadia, G.J.; Shukla, Y.N.; Chowdhury, B.K.; Basan,S.P.; Fales,

H.M.; Sokoloski, E.A. J. Chem. Soc., Chem. Commun. 1977, 535.

228. Kapadia, G.J.; Shukla, Y.N.; Basak, S.P.; Sokoloski, E.A.; Fales,

H.M. Tetrahedron 1980, 36, 2441.

229. Kapadia, G.J.; Shukla, Y.N. Planta Med. 1993, 59, 568.

230. (a) Grafe, U.; Radics, L. J. Antibiotics 1986, 39, 162; (b) Graefe, U.;

Schade, W.; Fleck, W. Ger (East) DD 241,749 1986. (CA

107:P216174k)

231. Breinholt, J.; Demuth, H.; Heide, M.; Jensen, G.W.; Moller, I.L.;

Nielsen, R.I.; Olsen, C.E.; Rosendahl, C.N. Acta Chem.

Scand.1996, 50, 443.

232. Yan, Y.; Li, G.; Wang, F.; Mao, W. Huadong Huagong Xueyuan

Xuebao 1992, 18, 192. (CA 118:127985k)

233. Somogyi, L. Bull. Chem. Soc. Jpn. 2001, 74, 873.

234. Da Silva, J. F. M.; Garden, S. J.; Pinto, A. C. J. Braz. Chem. Soc.

2001, 12(3), 273.

235. Elliott, J.; Gardner, D. L. Anal. Biochem. 1976, 70, 268.

236. Palfi, G.; Palfi, Z. Mydica 1982, 27, 107.

237. Eriksen, A. B. Medd. Nor. Inst. Skogforsk. , 1976, 32, 389.

238. Shah, A.; Rahman, S. S.; de Biasi, V.; Camilleri, P. Anal. Commun.

1997, 34(11), 325.

Page 345: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

328

239. Chiyanzu, I.; Clarkson, C.; Smith, P. J.; Lehman, J.; Gut, J.;

Rosenthal, P. J.; Chibale, K. Bioorg. Med. Chem. 2005, 13, 3249.

240. Pandeya, S. N.; Sriram, D. Acta. Pharm. Turc. 1998, 40, 33.

241. Sarangapani, M.; Reddy, V. M. Indian J. Heterocycl.Chem. 1994, 3,

257.

242. Varma, R. S.; Nobles, W. L. J. Pharm. Sci. 1975, 64, 881.

243. Sridhar, S. K.; Pandeya, S. N.; Stables, J. P.; Ramesh, A. Eur. J. Med.

Chem. 2002, 16, 129.

244. Varma, M.; Pandeya, S, N.; Singh, K. N.; Stables, J. P. Acta Pharm.

2004, 54, 49.

245. Pandeya, S. N.; Yogeeswari, P.; Sriram, D.; De Clercq, E.;

Pannecouque, C.; Witvrouw, M. Chemotherapy 1999, 45, 192.

246. Pandeya, S. N.; Sriram, D.; Nath, G.; De Clercq, E. Eur. J. Med.

Chem. 2000, 35, 249.

247. Pandeya, S. N.; Sriram, D.; Nath, G.; De Clercq, E. Arzneim

Forsch./Drug Res. 2000, 50, 55.

248. Pandeya, S. N.; Yogeeswari, P.; Sriram, D.; Nath, G. Bull. Chim.

Farm.1998, 137, 321.

249. Pandeya, S. N.; Sriram, D.; Nath, G.; De Clercq, E. Farmaco

1999, 54, 624.

250. Pandeya, S. N.; Sriram, D.; Nath, G.; De Clercq, E. S. Indian J.

Pharm. Sci. 1999, 61, 358.

251. Pandeya, S. N.; Sriram, D.; Nath, G.; De Clercq, E. Pharm. Acta

Helv. 1999, 74, 11.

252. Pandeya, S. N.; Sriram, D.; Nath, G.; De Clercq, E. Eur. J. Pharm. Sci.

1999, 9, 25.

253. Singh, S. P.; Shukla, S. K.; Awasthi, L. P. Curr. Sci. 1983, 52,

766.

254. Marcu, G. Chimica complecsilor coordinative; Ed. Academiei

Bucuresti: Bucarest, 1984; 44.

255. Cerchiaro, G.; Micke, G. A.; Tavares, M. F. M.; Ferriera, A. M. D. C. J.

Mol. Catal. A: Chem, 2004, 221, 29.

256. Takeuchi, T.; Bottcher, A.; Quezada, C. M.; Simon, M. I.; Meade, T. J.;

Gray, H. B. J. Am. Chem. Soc. 1998, 120, 8555.

Page 346: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

329

257. Bacchi, A.; Carcelli, M.; Pelagatti, P.; Pelizzi, G.; Rodriguez- Arguelles,

M. C.; Rogolino, D.;Solinas, C.; Zani, F. J. Inorg. Biochem. 2005,

99, 397.

258. Cerchiaro, G.; Aquilano, K.; Filomeni, G.; Rotilio, G.; Ciriolo, M. R.;

Ferriera, A. M. D. C. J. Inorg. Biochem. 2005, 99, 1433.

259. Hibino, S.; Choshi, T. Nat. Prod. Rep. 2002, 19, 148.

260. Somei, M.; Yamada, F. Nat. Prod. Rep. 2003, 20, 216.

261. Howard, H. R. ; Lowe J. A.; Seeger, T. F.; Seymour, P. A.; Zorn, S.

H.; Maloney, P. R.; Ewing, F. E.; Newman, M. E.; Schmidt; A. W.;

Furman, J. S. ; Robinson, G. L. ; Jackson, E. ; Johnson, C.; Morrone,

J. J. Med. Chem. 1996, 29, 143.

262. Haynes, J.; Obiako, B.; Babal, P.; Stevens, T.; Am. J. Physiol.

Heart Circul. Physiol, 1999, 276, 1877.

263. Liu, Y. ; Liu, D. ; Printzenhoff, D. ; Coghlan, M. J. ; Harris, R. ;

Krafte, D. S. Eur. J. Pharmacol. 2002, 435, 153.

264. Maggio, R.; Scarselli, M.; Novi, F.; Millan, M. J.; Corsini, G. U. J.

Neurochem. 2003, 87, 631.

265. Kohno, J. ; Koguchi, Y. ; Nishio, M. ; Nakao, K. ; Kuroda, M. ;

Shimizu, R. ; Ohnuki, T. ; Komatsubara, S. J. Org. Chem. 2000, 65,

990.

266. Fréchard, A.; Fabre, N.; Péan, C.; Montaut, S.; Fauvel, M.-T. ;

Rollin, P.; Fourasté, I. Tetrahedron Letters, 2001, 42, 9015.

267. Monde, K. ; Sasaki, K. ; Shirata, A.; Tagusuki, M. Phytochemistry

1991, 30, 2915.

268. Kamano, Y. ; Zhang, H.-P.; Ichihara, Y. ; Kizu, H. ; Komiyama, K. ;

Pettit, G. R. Tetrahedron Lett. 1995, 36, 2783.

269. Balk-Bindseil, W. ; Helmke, E. ; Weyland, H. ; Laatsch, H. Liebigs

Ann. Chem. 1995, 1291.

270. Rasmussen, H. B.; MacLeod, J. K. J.Nat. Prod. 1997, 60, 1152.

271. Kawasaki, T. ; Nagaoka, M. ; Satoh, T. ; Okamoto, A. ; Ukon, R. ;

Ogawa, A. Tetrahedron 2004, 60, 3493.

272. Monde, K.; Sasaki, K.; Shirata, A.; Takasugi, M. Phytochemistry

1991, 30, 2915.

Page 347: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

330

273. Hashiba, I.; Ando, Y.; Kawakami, I.; Sakota, R.; Nagano, K.; Mori, T.

Jpn. Kokai Tokkyo Koho 7970,265 1979. (CA 91:P175191u)

274. Ono, Y.; Nishimura, F.; Tamaki, K.; Fujii, K. Jpn. Kokai Tokkyo

Koho 79 151,963 1979. (CA 93:P8016a)

275. Wenkert, E.; Bringi, N.V.; Choulett, H.E. Acta Chem. Scand. 1982,

36B, 348.

276. Kadin, S.B. US 4,730,004 1988. (CA 110:P23729y)

277. Holmes, R.E.; Jourdan, G.P. U.S. Publ. Pat. Appl. B 427,946

1976. (CA 85:P46381h)

278. Zhong, T. Huaxue Tongbao 1986, 35. (CA 105:171460r)

279. Kuo, L.H.; Hsu, J.P.; Chen, C.T. US 5973165 1999.

280. Igarashi, R.; Nakamura, A. Jpn. Kokai Tokkyo Koho JP

07,196,610 1995. (CA 123:P285775z)

281. Crestini, C.; Saladino, R. Synth. Commun. 1994, 24, 2835.

282. Soriano, D.S. J.Chem. Educ. 1993, 70, 332.

283. Colgan, S.T.; Pollard, E.B. J. Chromatogr. Sci. 1991, 29, 433.

284. Bergman, J.; Stalhandske, C. Tetrahedron Lett. 1994, 35, 5279.

285. Papageorgiou, C.; Borer, X. Helv. Chim. Acta. 1988, 71, 1079.

286. Isukura, S.K.K. Jpn. Kokai Tokkyo Koho 61 07,254 ,1986. (CA

105:P24186c)

287. Minami, T.; Matsumoto, M.; Agawa, T. J. Chem. Soc., Chem.

Commun. 1976,1053.

288. Minami, T.; Matsuzaki, N.; Ohshiro, Y.; Agawa, T. J. Chem. Soc.,

Perkin Trans. 1, 1980, 1731.

289. El-Kateb, A.A.; Hennawy, I.T.; Shabana, R.; Osman, F.H. Phosph.

Sulf. 1984, 20, 329.

290. Sichuan Institute of Tradicional Chinese Medicine Zhongcaoyao

1981, 12, 499. (CA 97:38796e).

291. Wu, K.; Zhang, M.; Fang, Z.; Huang, L. Yaoxue Xuebao 1985, 20, 821.

(CA 106:84327e).

292. Gu, Y.C.; Li, G.L.; Yang, Y.P.; Fu, J.P.; Li, C.Z. Yaoxue Xuebao

1989, 24, 629. (CA 112:178548d).

293. Pfeiffer, G.; Bauer, H. Liebigs Ann. Chem. 1980, 564.

Page 348: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

331

294. Banerji, K.D.; Mazumder, A.K.D.; Guha, S.K. J. Indian Chem.

Soc. 1976, 53, 923.

295. Grosjean, D. Salmon, L.G.; Cass, G.R. Environ. Sci. Technol.

1992, 26, 952.

296. Rucker, J.W.; Freeman, H.S.; Hsu, W.N. Text. Chem. Color. 1992, 24,

66.

297. Reidies, A.H.; Jensen, D.; Guisti, M. Text. Chem. Color. 1992, 24, 26.

298. Matsui, M.; Morita, Shibata, K.; Takase, Y. Nippon Kagaku Kaishi.

1982, 1268. (CA 97:114736c).

299. Jonnalagadda, S.B.; Simoyi, R.; Muthakia, G.K. J. Chem. Soc.,

Perkin Trans. 2, 1988, 1111.

300. Nikokavouras, J.; Vassilopoulos, G. Monatsh. Chem. 1981, 112,

1239.

301. Amat-Guerri, F.; López-González, M.M.C.; Maretinez-Utrilla, R.

Tetrahedron Lett. 1983, 24, 3749.

302. Cheung, W. S.; Patch, R. J.; Player; M. R. J. Org. Chem., 2005, 70 (9),

3741.

303. Manley, J. M.; Kalman, M. J.; Conway, B. G.; Ball, C. C.; Havens,

J. L.; Vaidyanathan, R. J. Org. Chem., 2003, 68 (16), 6447.

304. Furniss, B.S.; Hannaford, A.J.; Smith, P.W.G.; Tatchell, A.R. “Vogel’s

Text Book of Practical Organic Chemistry” 5th Edition, Longman

Scientific and Technical, Co-published in the United States with John

Wiley and Sons Inc., New York, 1991, 1077.

305. Somerai, T.; Szilagyi, L.; Horvath, S. Arch. Pharm., 1986, 139,

238.

306. H. Gilman and A. H. Blatt. Organic synthesis collective. 1967, Vol. 1, 2nd ed, J. Willey, New York, pp. 447.

307. Iqbal, R.; Rama, N. H.; Ahmad, N.; Zamani, K H. J. Chem. Soc.

Pak., 1997, 19, 77.

308. Iqbal, R.; Zamani, K. H.; Rama, N. H. Tr. J. of Chem., 1996, 20,

295.

309. Ozturk, S., Akkurt, M., Cansız, A., Koparır, M., S¸ ekerci, M. &

Heinemann,F. W.. Acta Cryst. 2004, E60, 425.

Page 349: SYNTHESIS, CHARACTERIZATION AND BIOLOGICAL ACTIVITIES OF

332

310. Zhang, L.-X., Zhang, A.-J., Lei, X.-X., Zou, K.-H. & Ng, S. W.

Acta Cryst. 2004, E60, 613.

311. Gauss, D. H.; Gruter, F.; Sprinzl, M. Nucl. Acid Res., 1979, 6, 49.